1
|
Chae H, Kim SY, Kang HM, Im SA, Youn YA. Dysbiosis of the initial stool microbiota increases the risk of developing necrotizing enterocolitis or feeding intolerance in newborns. Sci Rep 2024; 14:24416. [PMID: 39424878 PMCID: PMC11489565 DOI: 10.1038/s41598-024-75157-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
Several perinatal factors influence the intestinal microbiome of newborns during the first days of life, whether during delivery or even in utero. These factors may increase the risk of developing necrotizing enterocolitis (NEC) by causing dysbiosis linked to a NEC-associated microbiota, which may also be associated with other gastrointestinal problems. The objective of our study was to evaluate the potential risks associated with microbial shifts in newborns with gastrointestinal symptoms and identify the intestinal microbiota of neonates at risk for NEC.During the study period, 310 preterm and term newborns' first passed meconium occurring within 72 h of birth were collected, and the microbiome was analyzed. We identified the risk factors in the NEC/FI group. Regarding microbiota, we compared the bacterial abundance between the NEC/FI group at the phylum and genus levels and explored the differences in the microbial composition of the 1st stool samples. A total of 14.8% (n = 46) of the infants were diagnosed with NEC or FI. In univariate analysis, the mean gestational age and birth weight were significantly lower in the NEC/FI group (p < 0.001). Prolonged rupture of membranes (PROM) > 18 h, chorioamnionitis, and histology were significantly higher in the NEC/FI group (p < 0.001). Multivariate analysis showed that gestational age (GA), prolonged membrane rupture (> 18 h), and early onset sepsis were consistently associated with an increased risk of NEC/FI. Infants diagnosed with NEC/FI exhibited a significantly lower abundance of Actinobacteria at the phylum level than the control group (p < 0.001). At the genus level, a significantly lower abundance of Streptococcus and Bifidobacterium which belong to the Actinobacteria phylum, was observed in the NEC/FI group (p < 0.001). Furthermore, the NEC/FI had significantly lower alpha diversities (Shannon Index,3.39 vs. 3.12; P = 0.044, respectively). Our study revealed that newborns with lower diversity and dysbiosis in their initial gut microbiota had an increased risk of developing NEC, with microbiota differences appearing to be associated with NEC/FI. Dysbiosis could potentially serve as a predictive marker for NEC- or GI-related symptoms.
Collapse
Affiliation(s)
- Hyojin Chae
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sae Yun Kim
- Department of Pediatrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Hyun Mi Kang
- Department of Pediatrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Soo-Ah Im
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young-Ah Youn
- Department of Pediatrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
2
|
Tripp P, Davis EC, Gurung M, Rosa F, Bode L, Fox R, LeRoith T, Simecka C, Seppo AE, Järvinen KM, Yeruva L. Infant Microbiota Communities and Human Milk Oligosaccharide Supplementation Independently and Synergistically Shape Metabolite Production and Immune Responses in Healthy Mice. J Nutr 2024; 154:2871-2886. [PMID: 39069270 PMCID: PMC11393170 DOI: 10.1016/j.tjnut.2024.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Multiple studies have demonstrated associations between the early-life gut microbiome and incidence of inflammatory and autoimmune disease in childhood. Although microbial colonization is necessary for proper immune education, it is not well understood at a mechanistic level how specific communities of bacteria promote immune maturation or drive immune dysfunction in infancy. OBJECTIVES In this study, we aimed to assess whether infant microbial communities with different overall structures differentially influence immune and gastrointestinal development in healthy mice. METHODS Germ-free mice were inoculated with fecal slurries from Bifidobacterium longum subspecies infantis positive (BIP) or B. longum subspecies infantis negative (BIN) breastfed infants; half of the mice in each group were also supplemented with a pool of human milk oligosaccharides (HMOs) for 14 d. Cecal microbiome composition and metabolite production, systemic and mucosal immune outcomes, and intestinal morphology were assessed at the end of the study. RESULTS The results showed that inoculation with a BIP microbiome results in a remarkably distinct microbial community characterized by higher relative abundances of cecal Clostridium senu stricto, Ruminococcus gnavus, Cellulosilyticum sp., and Erysipelatoclostridium sp. The BIP microbiome produced 2-fold higher concentrations of cecal butyrate, promoted branched short-chain fatty acid (SCFA) production, and further modulated serotonin, kynurenine, and indole metabolism relative to BIN mice. Further, the BIP microbiome increased the proportions of innate and adaptive immune cells in spleen, while HMO supplementation increased proliferation of mesenteric lymph node cells to phorbol myristate acetate and lipopolysaccharide and increased serum IgA and IgG concentrations. CONCLUSIONS Different microbiome compositions and HMO supplementation can modulate SCFA and tryptophan metabolism and innate and adaptive immunity in young, healthy mice, with potentially important implications for early childhood health.
Collapse
Affiliation(s)
- Patricia Tripp
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States
| | - Manoj Gurung
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Fernanda Rosa
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, United States; Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Renee Fox
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Tanya LeRoith
- Department of Biomedical Sciences & Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Christy Simecka
- Division of Laboratory Animal Medicine University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States.
| | - Laxmi Yeruva
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States.
| |
Collapse
|
3
|
Davis EC, Monaco CL, Insel R, Järvinen KM. Gut microbiome in the first 1000 days and risk for childhood food allergy. Ann Allergy Asthma Immunol 2024; 133:252-261. [PMID: 38494114 PMCID: PMC11344696 DOI: 10.1016/j.anai.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
OBJECTIVE To summarize recent data on the association between gut microbiome composition and food allergy (FA) in early childhood and highlight potential host-microbiome interactions that reinforce or abrogate oral tolerance. DATA SOURCES PubMed search of English-language articles related to FA, other atopic disease, and the gut microbiome in pregnancy and early childhood. STUDY SELECTIONS Human studies published after 2015 assessing the relationship between the gut bacteriome and virome in the first 2 years of life and FA or food sensitization development in early childhood were prioritized. Additional human studies conducted on the prenatal gut microbiome or other atopic diseases and preclinical studies are also discussed. RESULTS Children who developed FA harbored lower abundances of Bifidobacterium and Clostridia species and had a less mature microbiome during infancy. The early bacterial microbiome protects against FA through production of anti-inflammatory metabolites and induction of T regulatory cells and may also affect FA risk through a role in trained immunity. Infant enteric phage communities are related to childhood asthma development, though no data are available for FA. Maternal gut microbiome during pregnancy is associated with childhood FA risk, potentially through transplacental delivery of maternal bacterial metabolites, though human studies are lacking. CONCLUSION The maternal and infant microbiomes throughout the first 1000 days of life influence FA risk through a number of proposed mechanisms. Further large, longitudinal cohort studies using taxonomic, functional, and metabolomic analysis of the bacterial and viral microbiomes are needed to provide further insight on the host-microbe interactions underlying FA pathogenesis in childhood.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Cynthia L Monaco
- Division of Infectious Disease, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Richard Insel
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| |
Collapse
|
4
|
Liu L, Chen C, Li Y, Ao D, Wu J, Cai N, Li W, Xiang M. Dynamics alteration of the gut microbiota and faecal metabolomes in very low or extremely low birth weight infants: a Chinese single-center, prospective cohort study. Front Microbiol 2024; 15:1438213. [PMID: 39247697 PMCID: PMC11377216 DOI: 10.3389/fmicb.2024.1438213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Objective The aim of this study is to comprehensively investigate the temporal dynamics of faecal gut microbiota and metabonomics in early postnatal with a focus on very low or extremely low birth weight (VLBW/ELBW) infants. Methods We collected faecal samples from 157 VLBW/ELBW infants at three time points: days 1, 14, and 28 in a prospective cohort study. The faecal microbial diversity, abundance, composition, and metabolomic analyses were determined using 16S rRNA sequencing and liquid chromatography tandem mass spectrometry (LC-MS/MS). Microbiome functional analyses were conducted utilizing PICRUSt2. The ecological association networks were employed to investigate the interactions between gut microbiota and identify the core genus within 28 days of birth, as well as to unveil correlations between taxa and metabolites. Result (1) The alpha diversity of gut microbiota significantly decreased from D1 to D28, accompanied by an interrupted trajectory lacking obligate anaerobes. At the phylum level, the 16S RNA sequencing results showed an increase in Proteobacteria and a decrease in Firmicutes and Bacteroidota from D1 to D28. At the genus level, there was a decrease in the relative abundance of Staphylococcus, Acinetobacter and Ureaplasma, with Klebsiella and Enterococcus emerging as the most abundant genera. (2) The analysis revealed a total of 561 metabolic markers that exhibited significant and distinct alterations between D1 and D14. (3) Ecological association networks revealed that the gut microbiota in D1 exhibited a significantly higher degree of microbial interactions compared to those in D14 and D28. Additionally, Enterococcus, Klebsiella, and Enterobacter were major contributors to the co-occurring network at these three time points. (4) Steroid hormone biosynthesis, including tetrahydrocortisone, androsterone glucuronide, androstenedione and etiocholanolone glucuronide, decreased within 28 days after birth. Conclusion We have successfully demonstrated a significant dysbiosis in the gut microbiota and a subsequent decrease in its diversity within 4 weeks postpartum in VLBW/ELBW infants. Monitoring the gut microbiota of VLBW/ELBW infants and promptly rectifying dysbiosis in the early stages may represent a potential therapeutic strategy.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chaohong Chen
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - YeShan Li
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Dang Ao
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiayuan Wu
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Nali Cai
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wen Li
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Min Xiang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
5
|
Roberts AG, Younge N, Greenberg RG. Neonatal Necrotizing Enterocolitis: An Update on Pathophysiology, Treatment, and Prevention. Paediatr Drugs 2024; 26:259-275. [PMID: 38564081 DOI: 10.1007/s40272-024-00626-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease predominantly affecting premature and very low birth weight infants resulting in inflammation and necrosis of the small bowel and colon and potentially leading to sepsis, peritonitis, perforation, and death. Numerous research efforts have been made to better understand, treat, and prevent NEC. This review explores a variety of factors involved in the pathogenesis of NEC (prematurity, low birth weight, lack of human breast milk exposure, alterations to the microbiota, maternal and environmental factors, and intestinal ischemia) and reports treatment modalities surrounding NEC, including pain medications and common antibiotic combinations, the rationale for these combinations, and recent antibiotic stewardship approaches surrounding NEC treatment. This review also highlights the effect of early antibiotic exposure, infections, proton pump inhibitors (PPIs), and H2 receptor antagonists on the microbiota and how these risk factors can increase the chances of NEC. Finally, modern prevention strategies including the use of human breast milk and standardized feeding regimens are discussed, as well as promising new preventative and treatment options for NEC including probiotics and stem cell therapy.
Collapse
|
6
|
Lenfestey MW, Li N, Gauthier J, Winglee K, Fodor A, Zeng K, Jobin C, Neu J, Parker LA. Effect of Routine Gastric Residual Aspiration on the Preterm Infant Fecal Microbiome. Am J Perinatol 2024; 41:e212-e220. [PMID: 35709728 DOI: 10.1055/a-1877-6306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
OBJECTIVE Enteral feeding tubes are used in neonatal intensive care units (NICUs) to assess feeding tolerance by utilizing preprandial gastric residual aspiration. This study evaluates the effect of gastric residual aspiration on the preterm infant fecal microbiome and gastrointestinal inflammation. STUDY DESIGN Fifty-one very low birth weight (VLBW) infants (≤32 weeks' gestational age and ≤1,250 g) enrolled in a larger single-center randomized controlled trial evaluating the effects of routine and nonroutine gastric residual aspiration were selected for further analysis. Of those infants, 30 had microbiome analysis performed on stools collected at 6 weeks by sequencing the bacterial V1 to V3 variable regions of the genes encoding for 16S rRNA. In an additional 21 infants, stool samples collected at 3 and 6 weeks were analyzed for intestinal inflammation using a cytokine multiplex panel. RESULTS Microbial communities between groups were not distinct from each other and there was no difference in intestinal inflammation between groups. Analyses using gene expression packages DESeq2 and edgeR produced statistically significant differences in several taxa, possibly indicating a more commensal intestinal microbiome in infants not undergoing gastric residual aspiration. CONCLUSION Omission of routine gastric residual aspiration was not associated with intestinal dysbiosis or inflammation, providing additional evidence that monitors preprandial gastric residuals is unnecessary. KEY POINTS · Omission of routine gastric residual aspiration was not associated with intestinal dysbiosis or inflammation.. · Existing literature indicates preprandial gastric aspiration does not reliably correlate with development of necrotizing enterocolitis but does correlate with delayed enteral nutrition.. · Further study is required but this data that suggest monitoring preprandial gastric residuals are unnecessary..
Collapse
Affiliation(s)
- Mary W Lenfestey
- Department of Pediatrics, Pediatric Gastroenterology, East Carolina University, Greenville, North Carolina
| | - Nan Li
- Department of Pediatrics, Division of Neonatology, University of Florida, Gainesville, Florida
| | - Josee Gauthier
- Division of Gastroenterology, Department of Medicine, University of Florida, Gainesville, Florida
| | - Kathryn Winglee
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina
| | - Anthony Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina
| | - Ke Zeng
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina
| | - Christian Jobin
- Department of Medicine, Gastroenterology, University of Florida, Gainesville, Florida
| | - Josef Neu
- Department of Pediatrics, Neonatology, University of Florida, Gainesville, Florida
| | - Leslie A Parker
- College of Nursing, University of Florida, Gainesville, Florida
| |
Collapse
|
7
|
Mortensen JS, Bohr SSR, Krog LS, Bøtker JP, Kapousidou V, Saaby L, Hatzakis NS, Mørck Nielsen H, Nguyen DN, Rønholt S. Neonatal intestinal mucus barrier changes in response to maturity, inflammation, and sodium decanoate supplementation. Sci Rep 2024; 14:7665. [PMID: 38561398 PMCID: PMC10985073 DOI: 10.1038/s41598-024-58356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
The integrity of the intestinal mucus barrier is crucial for human health, as it serves as the body's first line of defense against pathogens. However, postnatal development of the mucus barrier and interactions between maturity and its ability to adapt to external challenges in neonatal infants remain unclear. In this study, we unveil a distinct developmental trajectory of the mucus barrier in preterm piglets, leading to enhanced mucus microstructure and reduced mucus diffusivity compared to term piglets. Notably, we found that necrotizing enterocolitis (NEC) is associated with increased mucus diffusivity of our large pathogen model compound, establishing a direct link between the NEC condition and the mucus barrier. Furthermore, we observed that addition of sodium decanoate had varying effects on mucus diffusivity depending on maturity and health state of the piglets. These findings demonstrate that regulatory mechanisms governing the neonatal mucosal barrier are highly complex and are influenced by age, maturity, and health conditions. Therefore, our results highlight the need for specific therapeutic strategies tailored to each neonatal period to ensure optimal gut health.
Collapse
Affiliation(s)
- Janni Støvring Mortensen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Søren S-R Bohr
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Department of Chemistry and Nanoscience Center, Faculty of Science, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Lasse Skjoldborg Krog
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Johan Peter Bøtker
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Vaya Kapousidou
- Department of Chemistry and Nanoscience Center, Faculty of Science, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Lasse Saaby
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Nikos S Hatzakis
- Department of Chemistry and Nanoscience Center, Faculty of Science, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
- NovoNordisk Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 68, 1870, Frederiksberg C, Denmark.
| | - Stine Rønholt
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| |
Collapse
|
8
|
Patangia DV, Grimaud G, O'Shea CA, Ryan CA, Dempsey E, Stanton C, Ross RP. Early life exposure of infants to benzylpenicillin and gentamicin is associated with a persistent amplification of the gut resistome. MICROBIOME 2024; 12:19. [PMID: 38310316 PMCID: PMC10837951 DOI: 10.1186/s40168-023-01732-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/24/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Infant gut microbiota is highly malleable, but the long-term longitudinal impact of antibiotic exposure in early life, together with the mode of delivery on infant gut microbiota and resistome, is not extensively studied. METHODS Two hundred and eight samples from 45 infants collected from birth until 2 years of age over five time points (week 1, 4, 8, 24, year 2) were analysed. Based on shotgun metagenomics, the gut microbial composition and resistome profile were compared in the early life of infants divided into three groups: vaginal delivery/no-antibiotic in the first 4 days of life, C-section/no-antibiotic in the first 4 days of life, and C-section/antibiotic exposed in first 4 days of life. Gentamycin and benzylpenicillin were the most commonly administered antibiotics during this cohort's first week of life. RESULTS Newborn gut microbial composition differed in all three groups, with higher diversity and stable composition seen at 2 years of age, compared to week 1. An increase in microbial diversity from week 1 to week 4 only in the C-section/antibiotic-exposed group reflects the effect of antibiotic use in the first 4 days of life, with a gradual increase thereafter. Overall, a relative abundance of Actinobacteria and Bacteroides was significantly higher in vaginal delivery/no-antibiotic while Proteobacteria was higher in C-section/antibiotic-exposed infants. Strains from species belonging to Bifidobacterium and Bacteroidetes were generally persistent colonisers, with Bifidobacterium breve and Bifidobacterium bifidum species being the major persistent colonisers in all three groups. Bacteroides persistence was dominant in the vaginal delivery/no-antibiotic group, with species Bacteroides ovatus and Phocaeicola vulgatus found to be persistent colonisers in the no-antibiotic groups. Most strains carrying antibiotic-resistance genes belonged to phyla Proteobacteria and Firmicutes, with the C-section/antibiotic-exposed group presenting a higher frequency of antibiotic-resistance genes (ARGs). CONCLUSION These data show that antibiotic exposure has an immediate and persistent effect on the gut microbiome in early life. As such, the two antibiotics used in the study selected for strains (mainly Proteobacteria) which were multiple drug-resistant (MDR), presumably a reflection of their evolutionary lineage of historical exposures-leading to what can be an extensive and diverse resistome. Video Abstract.
Collapse
Affiliation(s)
- Dhrati V Patangia
- School of Microbiology, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy Co., Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Ghjuvan Grimaud
- Teagasc Food Research Centre, Moorepark, Fermoy Co., Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | | | - C A Ryan
- APC Microbiome Ireland, Cork, Ireland
| | - Eugene Dempsey
- APC Microbiome Ireland, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- Infant Research Centre, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy Co., Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, Cork, Ireland.
| |
Collapse
|
9
|
Turunen J, Tejesvi MV, Paalanne N, Pokka T, Amatya SB, Mishra S, Kaisanlahti A, Reunanen J, Tapiainen T. Investigating prenatal and perinatal factors on meconium microbiota: a systematic review and cohort study. Pediatr Res 2024; 95:135-145. [PMID: 37591927 PMCID: PMC10798900 DOI: 10.1038/s41390-023-02783-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/30/2023] [Accepted: 07/29/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND The first-pass meconium has been suggested as a proxy for the fetal gut microbiota because it is formed in utero. This systematic review and cohort study investigated how pre- and perinatal factors influence the composition of the meconium microbiota. METHODS We performed the systematic review using Covidence by searching PubMed, Scopus, and Web of Science databases with the search terms "meconium microbiome" and "meconium microbiota". In the cohort study, we performed 16 S rRNA gene sequencing on 393 meconium samples and analyzed the sequencing data using QIIME2. RESULTS Our systematic review identified 69 studies exploring prenatal factors, immediate perinatal factors, and microbial composition in relation to subsequent health of infants but gave only limited comparative evidence regarding factors related to the composition of the meconium microbiota. The cohort study pointed to a low-biomass microbiota consisting of the phyla Firmicutes, Proteobacteria and Actinobacteriota and the genera Staphylococcus, Escherichia-Shigella and Lactobacillus, and indicated that immediate perinatal factors affected the composition of the meconium microbiota more than did prenatal factors. CONCLUSIONS This finding supports the idea that the meconium microbiota mostly starts developing during delivery. IMPACT It is unclear when the first-pass meconium microbiota develops, and what are the sources of the colonization. In this systematic review, we found 69 studies exploring prenatal factors, immediate perinatal factors, and microbial composition relative to subsequent health of infants, but there was no consensus on the factors affecting the meconium microbiota development. In this cohort study, immediate perinatal factors markedly affected the meconium microbiota development while prenatal factors had little effect on it. As the meconium microbiota composition was influenced by immediate perinatal factors, the present study supports the idea that the initial gut microbiota develops mainly during delivery.
Collapse
Affiliation(s)
- Jenni Turunen
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland.
- Biocenter Oulu, University of Oulu, Oulu, Finland.
| | - Mysore V Tejesvi
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Ecology and Genetics, Faculty of Science, University of Oulu, Oulu, Finland
| | - Niko Paalanne
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Tytti Pokka
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Research Service Unit, Oulu University Hospital, Oulu, Finland
| | - Sajeen Bahadur Amatya
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Surbhi Mishra
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Anna Kaisanlahti
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Justus Reunanen
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Terhi Tapiainen
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
10
|
Bosco A, Piu C, Picciau ME, Pintus R, Fanos V, Dessì A. Metabolomics in NEC: An Updated Review. Metabolites 2023; 14:14. [PMID: 38248817 PMCID: PMC10821135 DOI: 10.3390/metabo14010014] [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: 11/11/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Necrotizing enterocolitis (NEC) represents the most common and lethal acute gastrointestinal emergency of newborns, mainly affecting those born prematurely. It can lead to severe long-term sequelae and the mortality rate is approximately 25%. Furthermore, the diagnosis is difficult, especially in the early stages, due to multifactorial pathogenesis and complex clinical pictures with mild and non-specific symptoms. In addition, the existing tests have poor diagnostic value. Thus, the scientific community has been focusing its attention on the identification of non-invasive biomarkers capable of prediction, early diagnosis and discriminating NEC from other intestinal diseases in order to intervene early and block the progression of the pathology. In this regard, the use of "omics" technologies, especially metabolomics and microbiomics, could be a fundamental synergistic strategy to study the pathophysiology of NEC. In addition, a deeper knowledge of the microbiota-host cross-talk can clarify the metabolic pathways potentially involved in the pathology, allowing for the identification of specific biomarkers. In this article, the authors analyze the state-of-the-art concerning the application of metabolomics and microbiota analysis to investigate this pathology and discuss the future possibility of the metabolomic fingerprint of patients for diagnostic purposes.
Collapse
Affiliation(s)
| | | | | | | | | | - Angelica Dessì
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, 09124 Cagliari, Italy; (A.B.); (C.P.); (M.E.P.); (R.P.); (V.F.)
| |
Collapse
|
11
|
Xiu W, Lin J, Hu Y, Tang H, Wu S, Yang C. Assessing multiple factors affecting the gut microbiome structure of very preterm infants. Braz J Med Biol Res 2023; 56:e13186. [PMID: 38088676 PMCID: PMC10712283 DOI: 10.1590/1414-431x2023e13186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
The composition and diversity of the gut microbiota are essential for the health and development of the immune system of infants. However, there is limited information on factors that influence the gut microbiota of very preterm infants. In this study, we analyzed factors that affect the gut microbiota of very preterm infants. The stool samples from 64 very preterm infants with a gestational age less than 32 weeks were collected for 16S rRNA gene sequencing. The infants were divided according to the delivery mode, antibiotic use during pregnancy, and feeding methods. The abundance of Proteobacteria was high in both cesarean (92.7%) and spontaneous (55.5%) delivery groups and then shifted to Firmicutes after the first week of birth. In addition, Proteobacteria was also the dominant phylum of infant gut microbiome for mothers with antibiotic use, with more than 50% after the first week of birth. In comparison, the dominant phylum for mothers without antibiotic use was Firmicutes. Proteobacteria level was also high in breastfeeding and mixed-feeding groups, consisting of more than 90% of the community. By contrast, Proteobacteria was the dominant phylum at the first week of birth but then shifted to Firmicutes for the formula-fed group. The alterations of gut microbiota in infants can affect their health condition during growth. This study confirmed that the different feeding types, delivery modes, and use of antibiotics during pregnancy can significantly affect the composition of the gut microbiota of very preterm infants.
Collapse
Affiliation(s)
- Wenlong Xiu
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jiajia Lin
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yanhua Hu
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Heng Tang
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Shuangchan Wu
- Institute of Medical Research, Northwestern Polytechnical University, Xian, Shanxi Province, China
| | - Changyi Yang
- Department of Neonatology, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
12
|
Liu L, Xiang M, Cai X, Wu B, Chen C, Cai N, Ao D. Multi-omics analyses of gut microbiota via 16S rRNA gene sequencing, LC-MS/MS and diffusion tension imaging reveal aberrant microbiota-gut-brain axis in very low or extremely low birth weight infants with white matter injury. BMC Microbiol 2023; 23:387. [PMID: 38057706 PMCID: PMC10699022 DOI: 10.1186/s12866-023-03103-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/02/2023] [Indexed: 12/08/2023] Open
Abstract
OBJECTIVE The goal of this study was to comprehensively investigate the characteristics of gut microbiota dysbiosis and metabolites levels in very low or extremely low birth weight (VLBW/ELBW) infants with white matter injury (WMI). METHODS In this prospective cohort study, preterm infants with gestational age < 32 weeks and weight < 1.5 kg were investigated. Additionally, fecal samples were collected on days zero, 14d and 28d after admission to the intensive care unit. All subjects underwent brain scan via MRI and DTI at a corrected gestational age of 37 ~ 40 weeks. Based on the results of MRI examination, the VLBW/ELBW infants were divided into two groups: WMI and non-WMI. Finally, based on a multi-omics approach, we performed 16S rRNA gene sequencing, LC-MS/MS, and diffusion tension imaging to identify quantifiable and informative biomarkers for WMI. RESULT We enrolled 23 patients with and 48 patients without WMI. The results of 16S RNA sequencing revealed an increase in the number of Staphylococcus and Acinetobacter species in the fecal samples of infants with WMI, as well as increasing levels of S. caprae and A._johnsonii. LEfSe analysis (LDA ≥ 4) showed that the WMI group carried an abundance of Staphylococcus species including S. caprae, members of the phyla Bacteroidota and Actinobacteriota, and Acinetobacter species. A total of 139 metabolic markers were significantly and differentially expressed between WMI and nWMI. KEGG pathway enrichment analysis revealed that the WMI group showed significant downregulation of 17 metabolic pathways including biosynthesis of arginine and primary bile acids. The WMI group showed delayed brain myelination, especially in the paraventricular white matter and splenium of corpus callosum. Staphylococcus species may affect WMI by downregulating metabolites such as cholic acid, allocholic acid, and 1,3-butadiene. Gut microbiota such as Acinetobacter and Bacteroidetes may alter white matter structurally by upregulating metabolites such as cinobufagin. CONCLUSION Based on 16S RNA sequencing results, severe gut microbiota dysbiosis was observed in the WMI group. The results might reveal damage to potential signaling pathways of microbiota-gut-brain axis in gut microbiota. The mechanism was mediated via downregulation of the bile acid biosynthetic pathway.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Min Xiang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xiangsheng Cai
- Guangzhou Cadre Health Management Center, Guangzhou Eleventh People's Hospital, Guangzhou, 510000, Guangdong, China
| | - Benqing Wu
- University of the Chinese Academy of Science-Shenzhen Hospital, Shenzhen, 518000, Guangdong, China
| | - Chaohong Chen
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Nali Cai
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Dang Ao
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China.
| |
Collapse
|
13
|
Stanikova A, Jouza M, Bohosova J, Slaby O, Jabandziev P. Role of the microbiome in pathophysiology of necrotising enterocolitis in preterm neonates. BMJ Paediatr Open 2023; 7:e002172. [PMID: 37918941 PMCID: PMC10626796 DOI: 10.1136/bmjpo-2023-002172] [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: 07/06/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
Although necrotising enterocolitis (NEC) is a serious, life-threatening disease, improved neonatal care is increasing the number of survivors with NEC among extremely preterm neonates. Therapy is nevertheless mostly symptomatic and the mortality rate remains high, especially among neonates requiring surgery. Therefore, it is important to focus on preventing the disease and modifiable risk factors. NEC's pathophysiology is multifaceted, with key factors being immaturity of the immune and barrier protective mechanisms of the premature gut and exaggerated proinflammatory reaction to insults like gut hypoxia, enteral nutrition or microbial dysbiosis. The role of the intestinal microbiome in the pathophysiology of NEC has been a subject of research for many years, but to date no specific pathogen or type of dysbiosis has been connected with NEC development. This review assesses current knowledge as to the role of the intestinal microbiota in the pathophysiology of NEC and the possibilities for positively influencing it.
Collapse
Affiliation(s)
- Andrea Stanikova
- Department of Neonatology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Jouza
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatrics, University Hospital Brno, Brno, Czech Republic
| | - Julia Bohosova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Biology, University Hospital Brno, Brno, Czech Republic
| | - Petr Jabandziev
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatrics, University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
14
|
Wala SJ, Sajankila N, Ragan MV, Duff AF, Wickham J, Volpe SG, Wang Y, Conces M, Dumbauld Z, Purayil N, Narayanan S, Rajab A, Mihi B, Bailey MT, Goodman SD, Besner GE. Superior performance of biofilm versus planktonic Limosilactobacillus reuteri in protection of the intestines and brain in a piglet model of necrotizing enterocolitis. Sci Rep 2023; 13:17740. [PMID: 37872187 PMCID: PMC10593788 DOI: 10.1038/s41598-023-44676-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of gastrointestinal-related death in premature infants. Its etiology is multifactorial, with intestinal dysbiosis playing a major role. Probiotics are a logical preventative therapy for NEC, however their benefits have been inconsistent. We previously developed a novel probiotic delivery system in which planktonic (free-living) Limosilactobacillus reuteri (Lr) is incubated with biocompatible dextranomer microspheres (DM) loaded with maltose (Lr-DM-maltose) to induce biofilm formation. Here we have investigated the effects of Lr-DM-maltose in an enteral feed-only piglet model of NEC. We found a significant decrease in the incidence of Definitive NEC (D-NEC), death associated with D-NEC, and activated microglia in the brains of piglets treated with Lr-DM-maltose compared to non-treated piglets. Microbiome analyses using 16S rRNA sequencing of colonic contents revealed a significantly different microbial community composition between piglets treated with Lr-DM-maltose compared to non-treated piglets, with an increase in Lactobacillaceae and a decrease in Clostridiaceae in Lr-DM-maltose-treated piglets. Furthermore, there was a significant decrease in the incidence of D-NEC between piglets treated with Lr-DM-maltose compared to planktonic Lr. These findings validate our previous results in rodents, and support future clinical trials of Lr in its biofilm state for the prevention of NEC in premature neonates.
Collapse
Affiliation(s)
- Samantha J Wala
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Nitin Sajankila
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Mecklin V Ragan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Audrey F Duff
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Joseph Wickham
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Samuel G Volpe
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Yijie Wang
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Miriam Conces
- Department of Pathology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Zachary Dumbauld
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nanditha Purayil
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Siddharth Narayanan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Adrian Rajab
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Belgacem Mihi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Michael T Bailey
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Steven D Goodman
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
| |
Collapse
|
15
|
Yeramilli V, Cheddadi R, Benjamin H, Martin C. The Impact of Stress, Microbial Dysbiosis, and Inflammation on Necrotizing Enterocolitis. Microorganisms 2023; 11:2206. [PMID: 37764050 PMCID: PMC10534571 DOI: 10.3390/microorganisms11092206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of intestinal morbidity and mortality in neonates. A large body of work exists; however, the pathogenesis of NEC remains poorly understood. Numerous predictors have been implicated in the development of NEC, with relatively less emphasis on maternal factors. Utilizing human tissue plays a crucial role in enhancing our comprehension of the underlying mechanisms accountable for this devastating disease. In this review, we will discuss how maternal stress affects the pathogenesis of NEC and how changes in the intestinal microbiome can influence the development of NEC. We will also discuss the results of transcriptomics-based studies and analyze the gene expression changes in NEC tissues and other molecular targets associated with the pathogenesis of NEC.
Collapse
Affiliation(s)
| | | | | | - Colin Martin
- Division of Pediatric, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. S., Lowder Building Suite 300, Birmingham, AL 35233, USA
| |
Collapse
|
16
|
Wala SJ, Ragan MV, Sajankila N, Volpe SG, Purayil N, Dumbauld Z, Besner GE. Probiotics and novel probiotic delivery systems. Semin Pediatr Surg 2023; 32:151307. [PMID: 37295299 DOI: 10.1016/j.sempedsurg.2023.151307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Necrotizing enterocolitis (NEC) is an infectious and inflammatory intestinal disease that is the most common surgical emergency in the premature patient population. Although the etiology of the disease is multifactorial, intestinal dysbiosis is a hallmark of this disease. Based on this, probiotics may play a therapeutic role in NEC by introducing beneficial bacteria with immunomodulating, antimicrobial, and anti-inflammatory functions into the gastrointestinal tract. Currently, there is no Food and Drug Administration (FDA)-approved probiotic for the prevention and treatment of NEC. All probiotic clinical studies to date have administered the bacteria in their planktonic (free-living) state. This review will discuss established probiotic delivery systems including planktonic probiotics, prebiotics, and synbiotics, as well as novel probiotic delivery systems such as biofilm-based and designer probiotics. We will also shed light on whether or not probiotic efficacy is influenced by administration with breast milk. Finally, we will consider the challenges associated with developing an FDA-approved probiotic for NEC.
Collapse
Affiliation(s)
- Samantha J Wala
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mecklin V Ragan
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nitin Sajankila
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Samuel G Volpe
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nanditha Purayil
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Zachary Dumbauld
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
17
|
Ragan MV, Wala SJ, Sajankila N, Duff AF, Wang Y, Volpe SG, Al-Hadidi A, Dumbauld Z, Purayil N, Wickham J, Conces MR, Mihi B, Goodman SD, Bailey MT, Besner GE. Development of a novel definitive scoring system for an enteral feed-only model of necrotizing enterocolitis in piglets. Front Pediatr 2023; 11:1126552. [PMID: 37138566 PMCID: PMC10149862 DOI: 10.3389/fped.2023.1126552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Necrotizing enterocolitis (NEC) is a complex inflammatory disorder of the human intestine that most often occurs in premature newborns. Animal models of NEC typically use mice or rats; however, pigs have emerged as a viable alternative given their similar size, intestinal development, and physiology compared to humans. While most piglet NEC models initially administer total parenteral nutrition prior to enteral feeds, here we describe an enteral-feed only piglet model of NEC that recapitulates the microbiome abnormalities present in neonates that develop NEC and introduce a novel multifactorial definitive NEC (D-NEC) scoring system to assess disease severity. Methods Premature piglets were delivered via Caesarean section. Piglets in the colostrum-fed group received bovine colostrum feeds only throughout the experiment. Piglets in the formula-fed group received colostrum for the first 24 h of life, followed by Neocate Junior to induce intestinal injury. The presence of at least 3 of the following 4 criteria were required to diagnose D-NEC: (1) gross injury score ≥4 of 6; (2) histologic injury score ≥3 of 5; (3) a newly developed clinical sickness score ≥5 of 8 within the last 12 h of life; and (4) bacterial translocation to ≥2 internal organs. Quantitative reverse transcription polymerase chain reaction was performed to confirm intestinal inflammation in the small intestine and colon. 16S rRNA sequencing was performed to evaluate the intestinal microbiome. Results Compared to the colostrum-fed group, the formula-fed group had lower survival, higher clinical sickness scores, and more severe gross and histologic intestinal injury. There was significantly increased bacterial translocation, D-NEC, and expression of IL-1α and IL-10 in the colon of formula-fed compared to colostrum-fed piglets. Intestinal microbiome analysis of piglets with D-NEC demonstrated lower microbial diversity and increased Gammaproteobacteria and Enterobacteriaceae. Conclusions We have developed a clinical sickness score and a new multifactorial D-NEC scoring system to accurately evaluate an enteral feed-only piglet model of NEC. Piglets with D-NEC had microbiome changes consistent with those seen in preterm infants with NEC. This model can be used to test future novel therapies to treat and prevent this devastating disease.
Collapse
Affiliation(s)
- Mecklin V. Ragan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Samantha J. Wala
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Nitin Sajankila
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Audrey F. Duff
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Yijie Wang
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Samuel G. Volpe
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Ameer Al-Hadidi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Zachary Dumbauld
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Nanditha Purayil
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Joseph Wickham
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Miriam R. Conces
- Department of Pathology, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Belgacem Mihi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
| | - Steven D. Goodman
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Michael T. Bailey
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Gail E. Besner
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatric Surgery, Nationwide Children’s Hospital, Columbus, OH, United States
| |
Collapse
|
18
|
Abstract
The last decade has witnessed a meteoric rise in research focused on characterizing the human microbiome and identifying associations with disease risk. The advent of sequencing technology has all but eradicated gel-based fingerprinting approaches for studying microbial ecology, while at the same time traditional microbiological culture is undergoing a renaissance. Although multiplexed high-throughput sequencing is relatively new, the discoveries leading to this are nearly 50 years old, coinciding with the inaugural Microbiology Society Fleming Prize lecture. It was an honour to give the 2022 Fleming Prize lecture and this review will cover the topics from that lecture. The focus will be on the bacterial community in early life, beginning with term infants before moving on to infants delivered prematurely. The review will discuss recent work showing how human milk oligosaccharides (HMOs), an abundant but non-nutritious component of breast milk, can modulate infant microbiome and promote the growth of Bifidobacterium spp. This has important connotations for preterm infants at risk of necrotizing enterocolitis, a devastating intestinal disease representing the leading cause of death and long-term morbidity in this population. With appropriate mechanistic studies, it may be possible to harness the power of breast milk bioactive factors and infant gut microbiome to improve short- and long-term health in infants.
Collapse
|
19
|
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
|
20
|
Nicolas CT, Carter SR, Martin CA. Impact of maternal factors, environmental factors, and race on necrotizing enterocolitis. Semin Perinatol 2023; 47:151688. [PMID: 36572622 DOI: 10.1016/j.semperi.2022.151688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Necrotizing enterocolitis (NEC) is a complex disease with a multifactorial etiology. As the leading cause of intestinal morbidity and mortality among premature infants, many resources are being dedicated to neonatal care and molecular targets in the newborn intestine. However, NEC is heavily influenced by maternal and perinatal factors as well. Given its nature, preventive approaches to NEC are more likely to improve outcomes than new treatment strategies. Therefore, this review focuses on maternal, environmental, and racial factors associated with the development of NEC, with an emphasis on those that may be modifiable to decrease the incidence of the disease.
Collapse
Affiliation(s)
- Clara T Nicolas
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stewart R Carter
- Department of Surgery, Division of Pediatric Surgery, University of Louisville School of Medicine, Louisville, KY, United States
| | - Colin A Martin
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States; Department of General and Thoracic Surgery, Children's of Alabama, Birmingham, AL, United States.
| |
Collapse
|
21
|
Necrotizing Enterocolitis: The Role of Hypoxia, Gut Microbiome, and Microbial Metabolites. Int J Mol Sci 2023; 24:ijms24032471. [PMID: 36768793 PMCID: PMC9917134 DOI: 10.3390/ijms24032471] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease that predominantly affects very low birth weight preterm infants. Development of NEC in preterm infants is accompanied by high mortality. Surgical treatment of NEC can be complicated by short bowel syndrome, intestinal failure, parenteral nutrition-associated liver disease, and neurodevelopmental delay. Issues surrounding pathogenesis, prevention, and treatment of NEC remain unclear. This review summarizes data on prenatal risk factors for NEC, the role of pre-eclampsia, and intrauterine growth retardation in the pathogenesis of NEC. The role of hypoxia in NEC is discussed. Recent data on the role of the intestinal microbiome in the development of NEC, and features of the metabolome that can serve as potential biomarkers, are presented. The Pseudomonadota phylum is known to be associated with NEC in preterm neonates, and the role of other bacteria and their metabolites in NEC pathogenesis is also discussed. The most promising approaches for preventing and treating NEC are summarized.
Collapse
|
22
|
Zhang X, Zhang Y, He Y, Zhu X, Ai Q, Shi Y. β-glucan protects against necrotizing enterocolitis in mice by inhibiting intestinal inflammation, improving the gut barrier, and modulating gut microbiota. J Transl Med 2023; 21:14. [PMID: 36627673 PMCID: PMC9830848 DOI: 10.1186/s12967-022-03866-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease with high morbidity and mortality, affecting preterm infants especially those with very low and extremely low birth weight. β-glucan has manifested multiple biological effects including anti-inflammatory, regulation of gut microbiota, and immunomodulatory activities. This study aimed to investigate the effects of β-glucan on NEC. METHODS Neonatal C57BL/6 mice were randomly divided into three groups: Control group, NEC group and β-glucan group. Newborn 3-day-old mice were gavaged with either 1 mg/ml β-glucan or phosphate buffer saline at 0.03 ml/g for 7 consecutive days before NEC induction and a NEC model was established with hypoxia combined with cold exposure and formula feeding. All the pups were killed after 72-h modeling. Hematoxylin-eosin staining was performed to assess the pathological injury to the intestines. The mRNA expression levels of inflammatory factors in intestinal tissues were determined using quantitative real-time PCR. The protein levels of TLR4, NF-κB and tight junction proteins in intestinal tissues were evaluated using western blotting and immunohistochemistry. 16S rRNA sequencing was performed to determine the structure of the gut microbiota. RESULTS β-glucan administration ameliorated intestinal injury of NEC mice; reduced the intestinal expression of TLR4, NF-κB, IL-1β, IL-6, and TNF-α; increased the intestinal expression of IL-10; and improved the expression of ZO-1, Occludin and Claudin-1 within the intestinal barrier. Pre-treatment with β-glucan also increased the proportion of Actinobacteria, Clostridium butyricum, Lactobacillus johnsonii, Lactobacillus murinus, and Lachnospiraceae bacterium mt14 and reduced the proportion of Klebsiella oxytoca g Klebsiella in the NEC model. CONCLUSION β-glucan intervention prevents against NEC in neonatal mice, possibly by suppressing the TLR4-NF-κB signaling pathway, improving intestinal barrier function, and partially regulating intestinal microbiota.
Collapse
Affiliation(s)
- Xingdao Zhang
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yuni Zhang
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xingwang Zhu
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qing Ai
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yuan Shi
- grid.488412.3Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
23
|
Zenner C, Chalklen L, Adjei H, Dalby MJ, Mitra S, Cornwell E, Shaw AG, Sim K, Kroll JS, Hall LJ. Noninvasive Fecal Cytokine and Microbiota Profiles Predict Commencement of Necrotizing Enterocolitis in a Proof-of-Concept Study. GASTRO HEP ADVANCES 2023; 2:666-675. [PMID: 37469521 PMCID: PMC10352139 DOI: 10.1016/j.gastha.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/01/2023] [Indexed: 07/21/2023]
Abstract
Background and Aims Necrotizing enterocolitis (NEC) is a life-threatening disease and the most common gastrointestinal emergency in premature infants. Accurate early diagnosis is challenging. Modified Bell's staging is routinely used to guide diagnosis, but early diagnostic signs are nonspecific, potentially leading to unobserved disease progression, which is problematic given the often rapid deterioration observed. We investigated fecal cytokine levels, coupled with gut microbiota profiles, as a noninvasive method to discover specific NEC-associated signatures that can be applied as potential diagnostic markers. Methods Premature babies born below 32 weeks of gestation were admitted to the 2-site neonatal intensive care unit (NICU) of Imperial College hospitals (St. Mary's or Queen Charlotte's & Chelsea) between January 2011 and December 2012. During the NICU stay, expert neonatologists grouped individuals by modified Bell's staging (healthy, NEC1, NEC2/3) and fecal samples from diapers were collected consecutively. Microbiota profiles were assessed by 16S rRNA gene amplicon sequencing and cytokine concentrations were measured by V-Plex multiplex assays. Results Early evaluation of microbiota profiles revealed only minor differences. However, at later time points, significant changes in microbiota composition were observed for Bacillota (adj. P = .0396), with Enterococcus being the least abundant in Bell stage 2/3 NEC. Evaluation of fecal cytokine levels revealed significantly higher concentrations of IL-1α (P = .045), IL-5 (P = .0074), and IL-10 (P = .032) in Bell stage 1 NEC compared to healthy individuals. Conclusion Differences in certain fecal cytokine profiles in patients with NEC indicate their potential use as diagnostic biomarkers to facilitate earlier diagnosis. Additionally, associations between microbial and cytokine profiles contribute to improving knowledge about NEC pathogenesis.
Collapse
Affiliation(s)
- Christian Zenner
- Intestinal Microbiome, School of Life Sciences, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Lisa Chalklen
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
| | - Helena Adjei
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
| | - Matthew J. Dalby
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
| | - Suparna Mitra
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Emma Cornwell
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - Alexander G. Shaw
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - Kathleen Sim
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - J. Simon Kroll
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - Lindsay J. Hall
- Intestinal Microbiome, School of Life Sciences, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, Germany
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
24
|
Hammond EN, Kates AE, Putman-Buehler N, Watson L, Godfrey JJ, Brys N, Deblois C, Steinberger AJ, Cox MS, Skarlupka JH, Haleem A, Bentz ML, Suen G, Safdar N. A quality improvement study on the relationship between intranasal povidone-iodine and anesthesia and the nasal microbiota of surgery patients. PLoS One 2022; 17:e0278699. [PMID: 36490265 PMCID: PMC9733847 DOI: 10.1371/journal.pone.0278699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The composition of the nasal microbiota in surgical patients in the context of general anesthesia and nasal povidone-iodine decolonization is unknown. The purpose of this quality improvement study was to determine: (i) if general anesthesia is associated with changes in the nasal microbiota of surgery patients and (ii) if preoperative intranasal povidone-iodine decolonization is associated with changes in the nasal microbiota of surgery patients. MATERIALS AND METHODS One hundred and fifty-one ambulatory patients presenting for surgery were enrolled in a quality improvement study by convenience sampling. Pre- and post-surgery nasal samples were collected from patients in the no intranasal decolonization group (control group, n = 54). Pre-decolonization nasal samples were collected from the preoperative intranasal povidone-iodine decolonization group (povidone-iodine group, n = 97). Intranasal povidone-iodine was administered immediately prior to surgery and continued for 20 minutes before patients proceeded for surgery. Post-nasal samples were then collected. General anesthesia was administered to both groups. DNA from the samples was extracted for 16S rRNA sequencing on an Illumina MiSeq. RESULTS In the control group, there was no evidence of change in bacterial diversity between pre- and post-surgery samples. In the povidone-iodine group, nasal bacterial diversity was greater in post-surgery, relative to pre-surgery (Shannon's Diversity Index (P = 0.038), Chao's richness estimate (P = 0.02) and Inverse Simpson index (P = 0.027). Among all the genera, only the relative abundance of the genus Staphylococcus trended towards a decrease in patients after application (FDR adjusted P = 0.06). Abundant genera common to both povidone-iodine and control groups included Staphylococcus, Bradyrhizobium, Corynebacterium, Dolosigranulum, Lactobacillus, and Moraxella. CONCLUSIONS We found general anesthesia was not associated with changes in the nasal microbiota. Povidone-iodine treatment was associated with nasal microbial diversity and decreased abundance of Staphylococcus. Future studies should examine the nasal microbiota structure and function longitudinally in surgical patients receiving intranasal povidone-iodine.
Collapse
Affiliation(s)
- Eric N. Hammond
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI, United States of America
- Division of Infectious Disease, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Ashley E. Kates
- Division of Infectious Disease, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States of America
| | - Nathan Putman-Buehler
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Lauren Watson
- SSM Health, St. Mary’s Hospital, Madison, WI, United States of America
| | - Jared J. Godfrey
- Division of Infectious Disease, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States of America
| | - Nicole Brys
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Courtney Deblois
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Andrew J. Steinberger
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Madison S. Cox
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Joseph H. Skarlupka
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Ambar Haleem
- Division of Infectious Disease, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Michael L. Bentz
- Division of Plastic and Reconstructive Surgery and Urology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Garret Suen
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Nasia Safdar
- Division of Infectious Disease, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States of America
- * E-mail:
| |
Collapse
|
25
|
Moschino L, Verlato G, Duci M, Cavicchiolo ME, Guiducci S, Stocchero M, Giordano G, Fascetti Leon F, Baraldi E. The Metabolome and the Gut Microbiota for the Prediction of Necrotizing Enterocolitis and Spontaneous Intestinal Perforation: A Systematic Review. Nutrients 2022; 14:nu14183859. [PMID: 36145235 PMCID: PMC9506026 DOI: 10.3390/nu14183859] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is the most devastating gastrointestinal emergency in preterm neonates. Research on early predictive biomarkers is fundamental. This is a systematic review of studies applying untargeted metabolomics and gut microbiota analysis to evaluate the differences between neonates affected by NEC (Bell’s stage II or III), and/or by spontaneous intestinal perforation (SIP) versus healthy controls. Five studies applying metabolomics (43 cases, 95 preterm controls) and 20 applying gut microbiota analysis (254 cases, 651 preterm controls, 22 term controls) were selected. Metabolomic studies utilized NMR spectroscopy or mass spectrometry. An early urinary alanine/histidine ratio >4 showed good sensitivity and predictive value for NEC in one study. Samples collected in proximity to NEC diagnosis demonstrated variable pathways potentially related to NEC. In studies applying untargeted gut microbiota analysis, the sequencing of the V3−V4 or V3 to V5 regions of the 16S rRNA was the most used technique. At phylum level, NEC specimens were characterized by increased relative abundance of Proteobacteria compared to controls. At genus level, pre-NEC samples were characterized by a lack or decreased abundance of Bifidobacterium. Finally, at the species level Bacteroides dorei, Clostridium perfringens and perfringens-like strains dominated early NEC specimens, whereas Clostridium butyricum, neonatale and Propionibacterium acnei those at disease diagnosis. Six studies found a lower Shannon diversity index in cases than controls. A clear separation of cases from controls emerged based on UniFrac metrics in five out of seven studies. Importantly, no studies compared NEC versus SIP. Untargeted metabolomics and gut microbiota analysis are interrelated strategies to investigate NEC pathophysiology and identify potential biomarkers. Expression of quantitative measurements, data sharing via biorepositories and validation studies are fundamental to guarantee consistent comparison of results.
Collapse
Affiliation(s)
- Laura Moschino
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
- Correspondence: ; Tel.: +39-049-821-3548
| | - Giovanna Verlato
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Miriam Duci
- Paediatric Surgery, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Maria Elena Cavicchiolo
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Silvia Guiducci
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Matteo Stocchero
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
- Laboratory of Mass Spectrometry and Metabolomics, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Giuseppe Giordano
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
- Laboratory of Mass Spectrometry and Metabolomics, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Francesco Fascetti Leon
- Paediatric Surgery, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
- Institute of Paediatric Research, Città della Speranza, Laboratory of Mass Spectrometry and Metabolomics, 35127 Padova, Italy
| |
Collapse
|
26
|
Wu S, Di S, Liu T, Shi Y. Emerging prediction methods for early diagnosis of necrotizing enterocolitis. Front Med (Lausanne) 2022; 9:985219. [PMID: 36186788 PMCID: PMC9523100 DOI: 10.3389/fmed.2022.985219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease of the digestive system that occurs in the neonatal period. NEC is difficult to diagnose early and the prognosis is poor. Previous studies have reported that abnormalities can be detected before the presentation of clinical symptoms. Based on an analysis of literature related to the early prediction of NEC, we provide a detailed review on the early prediction and diagnosis methods of NEC, including ultrasound, near-infrared spectroscopy, biomarkers, and intestinal microbiota. This review aimed to provide a reference for further research and clinical practice.
Collapse
|
27
|
Liu XC, Du TT, Gao X, Zhao WJ, Wang ZL, He Y, Bao L, Li LQ. Gut microbiota and short-chain fatty acids may be new biomarkers for predicting neonatal necrotizing enterocolitis: A pilot study. Front Microbiol 2022; 13:969656. [PMID: 36060739 PMCID: PMC9428482 DOI: 10.3389/fmicb.2022.969656] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDysbacteriosis is thought to play an important role in the pathogenesis of necrotizing enterocolitis (NEC). We aimed to identify new biomarkers among gut microbiota and short-chain fatty acids (SCFAs) for the early prediction of NEC.Materials and methodsThirty-four preterm infants with gestational ages of ≤ 34 weeks who developed gastrointestinal symptoms were divided into the NEC group (n = 17) and non-NEC group (n = 17). In the NEC group, the gut microbiota and SCFAs in feces were assessed when the infants were enrolled (Group P) and when they were diagnosed with NEC (Group N). In the non-NEC group, samples were assessed when the infants were enrolled (Group C).ResultsThe Ace and Chao1 indices were higher in Group P than in Group C (P < 0.05), and there was no difference between Groups C and N or between Groups P and N (P > 0.05). There was no significant difference in the Simpson and Shannon indices among Groups C, P and N (P > 0.05). The four main phyla showed no differences (P > 0.05) in composition, while at the genus level, compared with Group C, in Group P, Clostridioides, Blautia and Clostridium_sensu_stricto_1 were increased, while Lactobacillus and Bifidobacterium were decreased (P < 0.05). At the species level, Streptococcus salivarius and Rothia mucilaginosa increased, while Bifidobacterium animals subsp. lactis decreased (P < 0.05). In Group N, at the genus level, Stenotrophomonas, Streptococcus and Prevotella increased (P < 0.05). Compared with those in Group C, the levels of acetic acid, propanoic acid and butyric acid decreased significantly in Groups P and N (P < 0.05), and the areas under the curves (AUCs) of these three SCFAs between groups C and P were 0.73, 0.70, and 0.68, respectively.ConclusionThe increase in Streptococcus salivarius and Rothia mucilaginosa and decrease in Bifidobacterium_animals_subsp._lactis, as well as the decrease in acetic, propionic and butyric acids, may help in the early prediction of NEC.
Collapse
|
28
|
A Paternal Fish Oil Diet Preconception Modulates the Gut Microbiome and Attenuates Necrotizing Enterocolitis in Neonatal Mice. Mar Drugs 2022; 20:md20060390. [PMID: 35736193 PMCID: PMC9230221 DOI: 10.3390/md20060390] [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: 05/20/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 12/05/2022] Open
Abstract
Epidemiology and animal studies suggest that a paternal history of toxicant exposure contributes to the developmental origins of health and disease. Using a mouse model, our laboratory previously reported that a paternal history of in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) increased his offspring’s risk of developing necrotizing enterocolitis (NEC). Additionally, our group and others have found that formula supplementation also increases the risk of NEC in both humans and mice. Our murine studies revealed that intervening with a paternal fish oil diet preconception eliminated the TCDD-associated outcomes that are risk factors for NEC (e.g., intrauterine growth restriction, delayed postnatal growth, and preterm birth). However, the efficacy of a paternal fish oil diet in eliminating the risk of disease development in his offspring was not investigated. Herein, reproductive-age male mice exposed to TCDD in utero were weaned to a standard or fish oil diet for one full cycle of spermatogenesis, then mated to age-matched unexposed females. Their offspring were randomized to a strict maternal milk diet or a supplemental formula diet from postnatal days 7–10. Offspring colon contents and intestines were collected to determine the onset of gut dysbiosis and NEC. We found that a paternal fish oil diet preconception reduced his offspring’s risk of toxicant-driven NEC, which was associated with a decrease in the relative abundance of the Firmicutes phylum, but an increase in the relative abundance of the Negativicutes class.
Collapse
|
29
|
Saeed NK, Al-Beltagi M, Bediwy AS, El-Sawaf Y, Toema O. Gut microbiota in various childhood disorders: Implication and indications. World J Gastroenterol 2022; 28:1875-1901. [PMID: 35664966 PMCID: PMC9150060 DOI: 10.3748/wjg.v28.i18.1875] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/08/2022] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota has a significant role in gut development, maturation, and immune system differentiation. It exerts considerable effects on the child's physical and mental development. The gut microbiota composition and structure depend on many host and microbial factors. The host factors include age, genetic pool, general health, dietary factors, medication use, the intestine's pH, peristalsis, and transit time, mucus secretions, mucous immunoglobulin, and tissue oxidation-reduction potentials. The microbial factors include nutrient availability, bacterial cooperation or antagonism, and bacterial adhesion. Each part of the gut has its microbiota due to its specific characteristics. The gut microbiota interacts with different body parts, affecting the pathogenesis of many local and systemic diseases. Dysbiosis is a common finding in many childhood disorders such as autism, failure to thrive, nutritional disorders, coeliac disease, Necrotizing Enterocolitis, helicobacter pylori infection, functional gastrointestinal disorders of childhood, inflammatory bowel diseases, and many other gastrointestinal disorders. Dysbiosis is also observed in allergic conditions like atopic dermatitis, allergic rhinitis, and asthma. Dysbiosis can also impact the development and the progression of immune disorders and cardiac disorders, including heart failure. Probiotic supplements could provide some help in managing these disorders. However, we are still in need of more studies. In this narrative review, we will shed some light on the role of microbiota in the development and management of common childhood disorders.
Collapse
Affiliation(s)
- Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Manama 12, Bahrain
- Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Busaiteen 15503, Bahrain
| | - Mohammed Al-Beltagi
- Department of Pediatrics, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Adel Salah Bediwy
- Department of Chest Disease, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
- Department of Pulmonology, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
| | - Yasser El-Sawaf
- Department of Tropical Medicine, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
- Department of Gastroenterology, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al-Habib Medical Group, Manama 26671, Bahrain
| | - Osama Toema
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| |
Collapse
|
30
|
Lin YC, Salleb-Aouissi A, Hooven TA. Interpretable prediction of necrotizing enterocolitis from machine learning analysis of premature infant stool microbiota. BMC Bioinformatics 2022; 23:104. [PMID: 35337258 PMCID: PMC8953333 DOI: 10.1186/s12859-022-04618-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/23/2022] [Indexed: 12/18/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a common, potentially catastrophic intestinal disease among very low birthweight premature infants. Affecting up to 15% of neonates born weighing less than 1500 g, NEC causes sudden-onset, progressive intestinal inflammation and necrosis, which can lead to significant bowel loss, multi-organ injury, or death. No unifying cause of NEC has been identified, nor is there any reliable biomarker that indicates an individual patient’s risk of the disease. Without a way to predict NEC in advance, the current medical strategy involves close clinical monitoring in an effort to treat babies with NEC as quickly as possible before irrecoverable intestinal damage occurs. In this report, we describe a novel machine learning application for generating dynamic, individualized NEC risk scores based on intestinal microbiota data, which can be determined from sequencing bacterial DNA from otherwise discarded infant stool. A central insight that differentiates our work from past efforts was the recognition that disease prediction from stool microbiota represents a specific subtype of machine learning problem known as multiple instance learning (MIL). Results We used a neural network-based MIL architecture, which we tested on independent datasets from two cohorts encompassing 3595 stool samples from 261 at-risk infants. Our report also introduces a new concept called the “growing bag” analysis, which applies MIL over time, allowing incorporation of past data into each new risk calculation. This approach allowed early, accurate NEC prediction, with a mean sensitivity of 86% and specificity of 90%. True-positive NEC predictions occurred an average of 8 days before disease onset. We also demonstrate that an attention-gated mechanism incorporated into our MIL algorithm permits interpretation of NEC risk, identifying several bacterial taxa that past work has associated with NEC, and potentially pointing the way toward new hypotheses about NEC pathogenesis. Our system is flexible, accepting microbiota data generated from targeted 16S or “shotgun” whole-genome DNA sequencing. It performs well in the setting of common, potentially confounding preterm neonatal clinical events such as perinatal cardiopulmonary depression, antibiotic administration, feeding disruptions, or transitions between breast feeding and formula. Conclusions We have developed and validated a robust MIL-based system for NEC prediction from harmlessly collected premature infant stool. While this system was developed for NEC prediction, our MIL approach may also be applicable to other diseases characterized by changes in the human microbiota. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04618-w.
Collapse
Affiliation(s)
- Yun Chao Lin
- Department of Computer Science, Columbia University, 1214 Amsterdam Ave., Mailcode 0401, New York, 10027, USA
| | - Ansaf Salleb-Aouissi
- Department of Computer Science, Columbia University, 1214 Amsterdam Ave., Mailcode 0401, New York, 10027, USA.
| | - Thomas A Hooven
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Richard King Mellon Institute for Pediatric Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| |
Collapse
|
31
|
Zioutis C, Seki D, Bauchinger F, Herbold C, Berger A, Wisgrill L, Berry D. Ecological Processes Shaping Microbiomes of Extremely Low Birthweight Infants. Front Microbiol 2022; 13:812136. [PMID: 35295290 PMCID: PMC8919028 DOI: 10.3389/fmicb.2022.812136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/07/2022] [Indexed: 11/23/2022] Open
Abstract
The human microbiome has been implicated in affecting health outcomes in premature infants, but the ecological processes governing early life microbiome assembly remain poorly understood. Here, we investigated microbial community assembly and dynamics in extremely low birth weight infants (ELBWI) over the first 2 weeks of life. We profiled the gut, oral cavity and skin microbiomes over time using 16S rRNA gene amplicon sequencing and evaluated the ecological forces shaping these microbiomes. Though microbiomes at all three body sites were characterized by compositional instability over time and had low body-site specificity (PERMANOVA, r 2 = 0.09, p = 0.001), they could nonetheless be clustered into four discrete community states. Despite the volatility of these communities, deterministic assembly processes were detectable in this period of initial microbial colonization. To further explore these deterministic dynamics, we developed a probabilistic approach in which we modeled microbiome state transitions in each ELBWI as a Markov process, or a "memoryless" shift, from one community state to another. This analysis revealed that microbiomes from different body sites had distinctive dynamics as well as characteristic equilibrium frequencies. Time-resolved microbiome sampling of premature infants may help to refine and inform clinical practices. Additionally, this work provides an analysis framework for microbial community dynamics based on Markov modeling that can facilitate new insights, not only into neonatal microbiomes but also other human-associated or environmental microbiomes.
Collapse
Affiliation(s)
- Christos Zioutis
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - David Seki
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Division of Neonatology, Department of Pediatrics and Adolescent Medicine, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Franziska Bauchinger
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Craig Herbold
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Department of Pediatrics and Adolescent Medicine, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Lukas Wisgrill
- Division of Neonatology, Department of Pediatrics and Adolescent Medicine, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - David Berry
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna, University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Vasilescu IM, Chifiriuc MC, Pircalabioru GG, Filip R, Bolocan A, Lazăr V, Diţu LM, Bleotu C. Gut Dysbiosis and Clostridioides difficile Infection in Neonates and Adults. Front Microbiol 2022; 12:651081. [PMID: 35126320 PMCID: PMC8810811 DOI: 10.3389/fmicb.2021.651081] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
In this review, we focus on gut microbiota profiles in infants and adults colonized (CDC) or infected (CDI) with Clostridioides difficile. After a short update on CDI epidemiology and pathology, we present the gut dysbiosis profiles associated with CDI in adults and infants, as well as the role of dysbiosis in C. difficile spores germination and multiplication. Both molecular and culturomic studies agree on a significant decrease of gut microbiota diversity and resilience in CDI, depletion of Firmicutes, Bacteroidetes, and Actinobacteria phyla and a high abundance of Proteobacteria, associated with low butyrogenic and high lactic acid-bacteria levels. In symptomatic cases, microbiota deviations are associated with high levels of inflammatory markers, such as calprotectin. In infants, colonization with Bifidobacteria that trigger a local anti-inflammatory response and abundance of Ruminococcus, together with lack of receptors for clostridial toxins and immunological factors (e.g., C. difficile toxins neutralizing antibodies) might explain the lack of clinical symptoms. Gut dysbiosis amelioration through administration of “biotics” or non-toxigenic C. difficile preparations and fecal microbiota transplantation proved to be very useful for the management of CDI.
Collapse
Affiliation(s)
- Iulia-Magdalena Vasilescu
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- INBI “Prof. Dr. Matei Balş” – National Institute for Infectious Diseases, Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- The Romanian Academy, Bucharest, Romania
- *Correspondence: Mariana-Carmen Chifiriuc,
| | | | - Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, Suceava, Romania
- Regional County Emergency Hospital, Suceava, Romania
| | - Alexandra Bolocan
- Department of General Surgery, University Emergency Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Veronica Lazăr
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Lia-Mara Diţu
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Coralia Bleotu
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Research Institute of the University of Bucharest, Bucharest, Romania
- Ştefan S. Nicolau Institute of Virology, Romanian Academy, Bucharest, Romania
| |
Collapse
|
33
|
Westaway JAF, Huerlimann R, Kandasamy Y, Miller CM, Norton R, Watson D, Infante-Vilamil S, Rudd D. To Probiotic or Not to Probiotic: A Metagenomic Comparison of the Discharge Gut Microbiome of Infants Supplemented With Probiotics in NICU and Those Who Are Not. Front Pediatr 2022; 10:838559. [PMID: 35345612 PMCID: PMC8957066 DOI: 10.3389/fped.2022.838559] [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: 12/17/2021] [Accepted: 02/09/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Preterm birth is associated with the development of both acute and chronic disease, and the disruption of normal gut microbiome development. Recent studies have sought to both characterize and understand the links between disease and the microbiome. Probiotic treatment may correct for these microbial imbalances and, in turn, mitigate disease. However, the criteria for probiotic supplementation in NICU's in North Queensland, Australia limits its usage to the most premature (<32 weeks gestation) and small for gestational age infants (<1,500 g). Here we use a combination of amplicon and shotgun metagenomic sequencing to compare the gut microbiome of infants who fulfill the criteria for probiotic-treatment and those who do not. The aims of this study were to determine if probiotic-supplemented preterm infants have significantly different taxonomic and functional profiles when compared to non-supplemented preterm infants at discharge. METHODS Preterm infants were recruited in North Queensland, Australia, with fecal samples collected just prior to discharge (36 ± 0.5 weeks gestation), to capture potential changes that could be probiotic induced. All samples underwent 16S rRNA gene amplicon sequencing, with a subset also used for shotgun metagenomics. Mixed effects models were used to assess the effect of probiotics on alpha diversity, beta diversity and taxonomic abundance, whilst accounting for other known covariates. RESULTS Mixed effects modeling demonstrated that probiotic treatment had a significant effect on overall community composition (beta diversity), characterized by greater alpha diversity and differing abundances of several taxa, including Bifidobacterium and Lactobacillus, in supplemented infants. CONCLUSION Late preterm-infants who go without probiotic-supplementation may be missing out on stabilizing-effects provided through increased alpha diversity and the presence of commensal microbes, via the use of probiotic-treatment. These findings suggest that late-preterm infants may benefit from probiotic supplementation. More research is needed to both understand the consequences of the differences observed and the long-term effects of this probiotic-treatment.
Collapse
Affiliation(s)
- Jacob A F Westaway
- College of Public Health, Medical and Veterinary Science, James Cook University, Cairns, QLD, Australia.,Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Townsville, QLD, Australia
| | - Roger Huerlimann
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Townsville, QLD, Australia.,Marine Climate Change Unit, Okinawa Institute of Science and Technology (OIST), Onna, Japan.,Center for Sustainable Tropical Fisheries and Aquaculture, James Cook University, Townsville, QLD, Australia
| | - Yoga Kandasamy
- College of Public Health, Medical and Veterinary Science, James Cook University, Townsville, QLD, Australia.,Neonatology, Townsville University Hospital, Townsville, QLD, Australia
| | - Catherine M Miller
- College of Public Health, Medical and Veterinary Science, James Cook University, Cairns, QLD, Australia
| | - Robert Norton
- Microbiology, Pathology Queensland, Herston, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - David Watson
- Maternal-Fetal Medicine, Townsville University Hospital, Townsville, QLD, Australia
| | - Sandra Infante-Vilamil
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Townsville, QLD, Australia.,Center for Sustainable Tropical Fisheries and Aquaculture, James Cook University, Townsville, QLD, Australia
| | - Donna Rudd
- College of Public Health, Medical and Veterinary Science, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
34
|
Spontaneous preterm delivery is reflected in both early neonatal and maternal gut microbiota. Pediatr Res 2022; 91:1804-1811. [PMID: 34349229 PMCID: PMC9270225 DOI: 10.1038/s41390-021-01663-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/22/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Aberrant gut microbiota composition in preterm neonates is linked to adverse health consequences. Little is known about the impact of perinatal factors or maternal gut microbiota on initial preterm gut colonization. METHODS Fecal samples were collected from 55 preterm neonates (<35 gestational weeks), 51 mothers, and 25 full-term neonates during the first 3-4 postpartum days. Gut microbiota composition was assessed using 16S ribosomal RNA gene sequencing. RESULTS Preterm neonates exhibited significantly lower gut microbiota alpha diversity and distinct beta diversity clustering compared to term neonates. Spontaneous preterm birth was associated with distinct initial gut microbiota beta diversity as compared to iatrogenic delivery. Gestational age or delivery mode had no impact on the preterm gut microbiota composition. The cause of preterm delivery was also reflected in the maternal gut microbiota composition. The contribution of maternal gut microbiota to initial preterm gut colonization was more pronounced after spontaneous delivery than iatrogenic delivery and not dependent on delivery mode. CONCLUSIONS The initial preterm gut microbiota is distinct from term microbiota. Spontaneous preterm birth is reflected in the early neonatal and maternal gut microbiota. Transmission of gut microbes from mother to neonate is determined by spontaneous preterm delivery, but not by mode of birth. IMPACT The initial gut microbiota in preterm neonates is distinct from those born full term. Spontaneous preterm birth is associated with changes in the gut microbiota composition of both preterm neonates and their mothers. The contribution of the maternal gut microbiota to initial neonatal gut colonization was more pronounced after spontaneous preterm delivery as compared to iatrogenic preterm delivery and not dependent on delivery mode. Our study provides new evidence regarding the early gut colonization patterns in preterm infants. Altered preterm gut microbiota has been linked to adverse health consequences and may provide a target for early intervention.
Collapse
|
35
|
Chang HY, Chiang Chiau JS, Ho YH, Chang JH, Tsai KN, Liu CY, Hsu CH, Lin CY, Ko MHJ, Lee HC. Impact of Early Empiric Antibiotic Regimens on the Gut Microbiota in Very Low Birth Weight Preterm Infants: An Observational Study. Front Pediatr 2021; 9:651713. [PMID: 34136438 PMCID: PMC8200535 DOI: 10.3389/fped.2021.651713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022] Open
Abstract
Frequent use of antibiotics in preterm infants disturbs their gut microbial balance. In this preliminary observational study, we investigated the effect of different antibiotic regimens, administered during the first week of life, on microbial composition and diversity in very low birth weight (VLBW) preterm infants. We performed fecal sampling of breastfed VLBW infants on days 7, 14, and 30. After excluding stool samples from infants who received probiotics or who were administered antibiotics beyond the age of 7 days, we compared gut microbiota profiles between infants receiving a combination of ampicillin and gentamicin for 3 days (AG group, n = 10) and those receiving a combination of ampicillin and cefotaxime for 7 days (AC group, n = 14) using 16S ribosomal DNA community profiling. We also assessed the changes over time in each group. Compared to the AG group, Enterococcus species were significantly more abundant in the AC group (P = 0.002), especially in 7-day samples (12.3 vs. 0.6%, respectively, P = 0.032). No difference was observed at phylum and genus level over time within each group. Species richness in the AC group decreased significantly in the 14-day (P = 0.038) and 30-day (P = 0.03) samples compared to that in the 7-day sample. The same was observed for microbial evenness; in contrast, no significant difference in Shannon index and beta-diversity was detected between the two groups. Controlling for relevant confounding variables did not change the results. In conclusion, different antibiotic regimens affect the early development of gut microbiota in VLBW preterm infants. Prolonged use of ampicillin and cefotaxime might result in overabundance of Enterococcus. However, given that no significant differences were observed in 1-month samples, bacterial genera appear to continue colonizing the gastrointestinal tract despite previous exposure to antibiotics. The clinical relevance of these findings should be elucidated by further studies.
Collapse
Affiliation(s)
- Hung-Yang Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | | | - Yu-Hsuan Ho
- Life Science, Delta Research Center, Delta Electronics Incorporation, Taipei, Taiwan
| | - Jui-Hsing Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Kun-Nan Tsai
- Life Science, Delta Research Center, Delta Electronics Incorporation, Taipei, Taiwan
| | - Chia-Yen Liu
- Life Science, Delta Research Center, Delta Electronics Incorporation, Taipei, Taiwan
| | - Chyong-Hsin Hsu
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
| | - Chia-Ying Lin
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
| | - Mary Hsin-Ju Ko
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
| | - Hung-Chang Lee
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| |
Collapse
|
36
|
de Cena JA, Zhang J, Deng D, Damé-Teixeira N, Do T. Low-Abundant Microorganisms: The Human Microbiome's Dark Matter, a Scoping Review. Front Cell Infect Microbiol 2021; 11:689197. [PMID: 34136418 PMCID: PMC8201079 DOI: 10.3389/fcimb.2021.689197] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/13/2021] [Indexed: 12/19/2022] Open
Abstract
Research on the human microbiome has mainly been restricted to the identification of most abundant microbiota associated with health or disease. Their abundance may reflect their capacity to exploit their niche, however, metabolic functions exerted by low-abundant microrganisms can impact the dysbiotic signature of local microbial habitats. This scoping review aims to map the literature regarding the management of low-abundant microorganisms in studies investigating human microbiome samples. A systematic literature search was performed in 5 electronic databases, as well as grey literature. We selected clinical microbiome studies targeting human participants of any age, from any body site. We also included studies with secondary data which originated from human biofilm samples. All of the papers used next-generation sequencing (NGS) techniques in their methodology. A total of 826 manuscripts were retrieved, of which 42 were included in this review and 22 reported low-abundant bacteria (LB) in samples taken from 7 body sites (breast, gut, oral cavity, skin, stomach, upper respiratory tract (URT), and vagina). Four studies reported microbes at abundance levels between 5 and 20%, 8 studies reported between 1 and 5%, and 18 studies reported below 1%. Fifteen papers mentioned fungi and/or archaea, and from those only 4 (fungi) and 2 (archaea) produced data regarding the abundance of these domains. While most studies were directed towards describing the taxonomy, diversity and abundance of the highly abundant species, low-abundant species have largely been overlooked. Indeed, most studies select a cut-off value at <1% for low-abundant organisms to be excluded in their analyses. This practice may compromise the true diversity and influence of all members of the human microbiota. Despite their low abundance and signature in biofilms, they may generate important markers contributing to dysbiosis, in a sort of ‘butterfly effect’. A detailed snapshot of the physiological, biological mechanisms at play, including virulence determinants in the context of a dysbiotic community, may help better understand the health-disease transition.
Collapse
Affiliation(s)
- Jéssica Alves de Cena
- Department of Dentistry, School of Health Sciences, University of Brasília, Brasilia, Brazil
| | - Jianying Zhang
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, Netherlands.,Xiangya School of Stomatology, Xiangya Stomatological Hospital, Central South University, Changsha, China
| | - Dongmei Deng
- Xiangya School of Stomatology, Xiangya Stomatological Hospital, Central South University, Changsha, China
| | - Nailê Damé-Teixeira
- Department of Dentistry, School of Health Sciences, University of Brasília, Brasilia, Brazil.,Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom
| | - Thuy Do
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
37
|
It's all in the milk: chondroitin sulfate as potential preventative therapy for necrotizing enterocolitis. Pediatr Res 2021; 89:1373-1379. [PMID: 32920604 PMCID: PMC8784957 DOI: 10.1038/s41390-020-01125-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 01/02/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating condition affecting up to 5% of neonatal intensive care unit (NICU) admissions. Risk factors include preterm delivery, low birth weight, and antibiotic use. The pathogenesis is characterized by a combination of intestinal ischemia, necrosis of the bowel, reperfusion injury, and sepsis typically resulting in surgical resection of afflicted bowel. Targeted medical therapy remains elusive. Chondroitin sulfate (CS) holds the potential to prevent the onset of NEC through its anti-inflammatory properties and protective effect on the gut microbiome. The purpose of this review is to outline the many properties of CS to highlight its potential use in high-risk infants and attenuate the severity of NEC. The purpose of this review is to (1) discuss the interaction of CS with the infant microbiome, (2) review the anti-inflammatory properties of CS, and (3) postulate on the potential role of CS in preventing NEC. IMPACT: NEC is a costly medical burden in the United States. Breast milk is the best preventative measure for NEC, but not all infants in the NICU have access to breast milk. Novel therapies and diagnostic tools are needed for NEC. CS may be a potential therapy for NEC due to its potent anti-inflammatory properties. CS could be added to the formula in an attempt to mitigate breast milk disparities.
Collapse
|
38
|
Cuna A, Morowitz MJ, Ahmed I, Umar S, Sampath V. Dynamics of the preterm gut microbiome in health and disease. Am J Physiol Gastrointest Liver Physiol 2021; 320:G411-G419. [PMID: 33439103 PMCID: PMC8238167 DOI: 10.1152/ajpgi.00399.2020] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Advances in metagenomics have allowed a detailed study of the gut microbiome, and its role in human health and disease. Infants born prematurely possess a fragile gut microbial ecosystem that is vulnerable to perturbation. Alterations in the developing gut microbiome in preterm infants are linked to life-threatening diseases such as necrotizing enterocolitis (NEC) and late-onset sepsis; and may impact future risk of asthma, atopy, obesity, and psychosocial disease. In this mini-review, we summarize recent literature on the origins and patterns of development of the preterm gut microbiome in the perinatal period. The host-microbiome-environmental factors that portend development of dysbiotic intestinal microbial patterns associated with NEC and sepsis are reviewed. Strategies to manipulate the microbiome and mitigate dysbiosis, including the use of probiotics and prebiotics will also be discussed. Finally, we explore the challenges and future directions of gut microbiome research in preterm infants.
Collapse
Affiliation(s)
- Alain Cuna
- 1Division of Neonatology, Children’s Mercy Kansas City, Kansas City, Missouri,2School of Medicine, University of Missouri Kansas City, Kansas City, Missouri
| | | | - Ishfaq Ahmed
- 4Department of Math, Science and Business Technology, Kansas City Kansas Community College, Kansas City, Kansas
| | - Shahid Umar
- 5Departments of Surgery and Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Venkatesh Sampath
- 1Division of Neonatology, Children’s Mercy Kansas City, Kansas City, Missouri,2School of Medicine, University of Missouri Kansas City, Kansas City, Missouri
| |
Collapse
|
39
|
Nguyen M, Holdbrooks H, Mishra P, Abrantes MA, Eskew S, Garma M, Oca CG, McGuckin C, Hein CB, Mitchell RD, Kazi S, Chew S, Casaburi G, Brown HK, Frese SA, Henrick BM. Impact of Probiotic B. infantis EVC001 Feeding in Premature Infants on the Gut Microbiome, Nosocomially Acquired Antibiotic Resistance, and Enteric Inflammation. Front Pediatr 2021; 9:618009. [PMID: 33665175 PMCID: PMC7921802 DOI: 10.3389/fped.2021.618009] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/08/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Preterm birth is a major determinant of neonatal survival and morbidity, but the gut microbiome and associated enteric inflammation are also key factors in neonatal development and the risk of associated morbidities. We prospectively and longitudinally followed two cohorts of preterm infants, one of which was fed activated Bifidobacterium longum subsp. infantis (B. infantis) EVC001 8 × 109 CFU daily, and the other was not fed a probiotic. Hospital feeding protocol assigned all infants born at <1500 g and/or < 32 weeks corrected gestational age to the probiotic feeding protocol, whereas infants born at >1500 g and/or >32 weeks corrected gestational age were not fed a probiotic. Fecal samples were opportunistically collected from 77 infants throughout the hospital stay, and subjected to shotgun metagenomic sequencing and quantification of enteric inflammation. De-identified metadata was collected from patient medical records. Results: The gut microbiome of preterm infants was typified by a high abundance of Enterobacteriaceae and/or Staphylococcaceae, and multivariate modeling identified the probiotic intervention, rather than degree of prematurity, day of life, or other clinical interventions, as the primary source of change in the gut microbiome. Among infants fed B. infantis EVC001, a high abundance of total Bifidobacteriaceae developed rapidly, the majority of which was B. infantis confirmed via subspecies-specific qPCR. Associated with this higher abundance of Bifidobacteriaceae, we found increased functional capacity for utilization of human milk oligosaccharides (HMOs), as well as reduced abundance of antibiotic resistance genes (ARGs) and the taxa that harbored them. Importantly, we found that infants fed B. infantis EVC001 exhibited diminished enteric inflammation, even when other clinical variables were accounted for using multivariate modeling. Conclusion: These results provide an important observational background for probiotic use in a NICU setting, and describe the clinical, physiological, and microbiome-associated improvements in preterm infants associated with B. infantis EVC001 feeding.
Collapse
Affiliation(s)
- Marielle Nguyen
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Heaven Holdbrooks
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Prasanthi Mishra
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Maria A. Abrantes
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Sherri Eskew
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Mariajamiela Garma
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Cyr-Geraurd Oca
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | | | | | | | - Sufyan Kazi
- Evolve Biosystems Inc., Davis, CA, United States
| | | | | | | | - Steven A. Frese
- Evolve Biosystems Inc., Davis, CA, United States
- Department of Food Science and Technology, University of Nebraska Lincoln, Lincoln, NE, United States
- Department of Nutrition, University of Nevada, Reno, NV, United States
| | - Bethany M. Henrick
- Evolve Biosystems Inc., Davis, CA, United States
- Department of Food Science and Technology, University of Nebraska Lincoln, Lincoln, NE, United States
| |
Collapse
|
40
|
Liu L, Ji M, Wang F, Tian Z, Yan Z, Wang S. N-acyl- l-homoserine lactones release and microbial community changes in response to operation temperature in an anammox biofilm reactor. CHEMOSPHERE 2021; 262:127602. [PMID: 32750589 DOI: 10.1016/j.chemosphere.2020.127602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 06/11/2023]
Abstract
A 1 L lab-scale anaerobic ammonium oxidation (anammox) biofilm reactor with nitrogen loading rate of 0.11 g/L d was run for 110 days with the operation temperature declining from 36 °C to 15 °C. The total inorganic nitrogen removal efficiency showed a reduction from 80% to 66%, when the temperature declined from 36 °C to 15 °C. N-acyl-l-homoserine lactones (AHLs) concentrations, especially C8-HSL and C6-HSL, declined in both water and biomass phases, and this decline indicated that the quorum sensing weakened. Microbial community analysis revealed that Candidatus Kuenenia was the predominant anammox bacteria during the entire operating period. The abundance of Candidatus Kuenenia increased from 1.43% to 22.89% when the temperature decreasing from 36 °C to 15 °C. The correlation between microbial genus and AHLs was complicated. Overall, the temperature decrease weakened the quorum sensing so that the nitrogen removal performance deteriorated, and increasing the anammox activity might be an efficient way to improve performance.
Collapse
Affiliation(s)
- Lingjie Liu
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Min Ji
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Fen Wang
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China.
| | - Zhongke Tian
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Zhao Yan
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Shuya Wang
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300350, China
| |
Collapse
|
41
|
Thänert R, Keen EC, Dantas G, Warner BB, Tarr PI. Necrotizing Enterocolitis and the Microbiome: Current Status and Future Directions. J Infect Dis 2020; 223:S257-S263. [PMID: 33330904 DOI: 10.1093/infdis/jiaa604] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Decades of research have failed to define the pathophysiology of necrotizing enterocolitis (NEC), a devastating pediatric gastrointestinal disorder of preterm infants. However, evidence suggests that host-microbiota interactions, in which microbial dysbiosis is followed by loss of barrier integrity, inflammation, and necrosis, are central to NEC development. Thus, greater knowledge of the preterm infant microbiome could accelerate attempts to diagnose, treat, and prevent NEC. In this article, we summarize clinical characteristics of and risk factors for NEC, the structure of the pre-event NEC microbiome, how this community interfaces with host immunology, and microbiome-based approaches that might prevent or lessen the severity of NEC in this very vulnerable population.
Collapse
Affiliation(s)
- Robert Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Eric C Keen
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA.,Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University in St Louis, St Louis, Missouri, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine in St Louis , St Louis, Missouri, USA
| | - Phillip I Tarr
- Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA.,Department of Pediatrics, Washington University School of Medicine in St Louis , St Louis, Missouri, USA
| |
Collapse
|
42
|
Chen K, Yuan T. The role of microbiota in neonatal hyperbilirubinemia. Am J Transl Res 2020; 12:7459-7474. [PMID: 33312382 PMCID: PMC7724329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/24/2020] [Indexed: 06/12/2023]
Abstract
Hyperbilirubinemia accounts for about 60% of full-term and 80% of preterm neonates globally, which is characterized by physiologically elevated unconjugated bilirubin in serum, but abnormally high levels of bilirubin have potential neurotoxic effects. Several factors contribute to the development of neonatal hyperbilirubinemia, including isoimmunization, dysregulated gut flora, genetic alteration and environmental factors. Animal studies have pinpointed the causal roles of several bacteria in bilirubin metabolism. Human studies have revealed microbiota composition in hyperbilirubinemia and found that gut microbiota affect newborns with different severity of hyperbilirubinemia. However, dysbiosis and subsequent changes in microbiota-related metabolic processes are not always considered. This review aims to describe the critical microbiota signatures for neonatal hyperbilirubinemia and focus on the underlying pathogenetic mechanism. These scientific bases give a new and accurate therapeutic strategy for the application of gut microbiota.
Collapse
Affiliation(s)
- Kewei Chen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health Zhejiang, PR China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health Zhejiang, PR China
| |
Collapse
|
43
|
Lu S, Huang Q, Wei B, Chen Y. Effects of β-Lactam Antibiotics on Gut Microbiota Colonization and Metabolites in Late Preterm Infants. Curr Microbiol 2020; 77:3888-3896. [PMID: 32970172 DOI: 10.1007/s00284-020-02198-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 09/03/2020] [Indexed: 12/19/2022]
Abstract
We usually refer to the critical period for intestinal flora establishment as infancy because the infant gut microbiota is characterized by low diversity and poor stability compared with that of adults. Moreover, it is also vulnerable to interference from a variety of factors. As β-lactam antibiotics are typically used in newborn infants with infectious diseases, we used 16S rDNA sequencing and LC-MS metabolomics to analyze fecal microbes and metabolites in 16 late preterm infants with or without β-lactam antibiotic treatment. The subjects were assigned to two groups: one not treated with antibiotics and another receiving β-lactam antibiotic treatment for less than seven days. Significant changes in fecal microbes and metabolites were observed in the late preterm infants treated with antibiotics, including a reduction in the diversity of the gut microbiota overall and some beneficial bacteria such as Bacteroides, whereas some opportunistic pathogenic bacteria such as Enterococcus showed an overgrowth trend. In addition, significant changes in some crucial metabolites were observed, such as amino acids and bile acids. These findings show that treatment with β-lactam antibiotics might affect the intestinal flora and its metabolites in late preterm infants in a short time period.
Collapse
Affiliation(s)
- Siliang Lu
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qingmei Huang
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Bingmei Wei
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yujun Chen
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
44
|
The microbiota protects against Pseudomonas aeruginosa pneumonia via γδ T cell-neutrophil axis in mice. Microbes Infect 2020; 22:294-302. [DOI: 10.1016/j.micinf.2020.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
|
45
|
Yu Y, Lu J, Oliphant K, Gupta N, Claud K, Lu L. Maternal administration of probiotics promotes gut development in mouse offsprings. PLoS One 2020; 15:e0237182. [PMID: 32764797 PMCID: PMC7413491 DOI: 10.1371/journal.pone.0237182] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022] Open
Abstract
Necrotizing enterocolitis is the most common gastrointestinal disorder in premature neonates. This disease is characterized by massive epithelial necrosis, gut barrier dysfunction and improper mucosal defense development. Studies have shown that probiotic administration can decrease NEC incidence and mortality. The proposed mechanisms of probiotics for the prevention of NEC are: promotion of intestinal development; improved barrier function through decreased apoptosis and improved mucin production; decreased expression of proinflammatory cytokines IL6, IL8, and TNFα, and modulation of microbiota dysbiosis in preterm infants. However, reported sepsis in the immunocompromised preterm host has deterred routine prophylactic administration of probiotics in the neonatal intensive care unit. We hypothesize that maternal administration of probiotics to pregnant mouse dams can recapitulate the beneficial effects observed in neonates fed with probiotics directly. We exposed pregnant mice to the probiotics and monitored the changes in the developing intestines of the offspring. Pregnant mice were fed daily with the probiotics Lactobacillus acidophilus and Bifidobacterium infantis (LB) from embryonic day15 to 2-week-old postnatally. Intraperitoneal administration of IL-1β in the pups was used to model proinflammatory insults. Sera were collected at 2 weeks of age and evaluated for inflammatory cytokines by enzyme-linked-immunosorbent-assay and gut permeability by Fluorescein isothiocyanate-dextran tracer assay. Ileal tissues were collected for the evaluation of apoptosis and proliferation of the intestinal epithelium; as well as mucin and tight junction integrity at mucosal surface by immunofluorescent staining. We find that maternal LB exposure facilitated intestinal epithelial cell differentiation, prevented loss of mucin and preserved the intestinal integrity and barrier function and decreased serum levels of IL-1β, TNF-α and IL-6 in the preweaned offsprings. in LB exposed pups. We demonstrate that maternal probiotic supplementation promotes gut maturation in developing offspring. This is potentially a safe alternative therapy to induce intestinal maturation and prevent prematurity-associated neonatal disorders.
Collapse
Affiliation(s)
- Yueyue Yu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Jing Lu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Kaitlyn Oliphant
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Nikhilesh Gupta
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Katerina Claud
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Lei Lu
- Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
46
|
Mokshagundam S, Ding T, Rumph JT, Dallas M, Stephens VR, Osteen KG, Bruner-Tran KL. Developmental 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure of either parent enhances the risk of necrotizing enterocolitis in neonatal mice. Birth Defects Res 2020; 112:1209-1223. [PMID: 32519502 DOI: 10.1002/bdr2.1742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/22/2020] [Accepted: 05/20/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a rare, but potentially fatal intestinal inflammatory condition most often arising in premature infants. Infants provided formula are also at greater risk of developing this disease. Although the majority of formula-fed, preterm infants do not develop NEC, up to 30% of infants with the disease do not survive. Thus, identifying additional, currently unrecognized factors, which may predispose a specific infant to NEC development would be a significant clinical advancement. In this regard, we have previously reported that offspring of female or male mice with a history of developmental exposure to the environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exhibit altered sensitivity to inflammatory challenges and are frequently born premature. Herein, we examined the possibility that, compared to unexposed mice (F1NONE ), developmental TCDD exposure of either parent (maternal, F1MTCDD , or paternal, F1PTCDD ) would enhance the risk of NEC in offspring (F2TCDD mice) in association with supplemental formula feeding. METHODS Beginning on postnatal day 7, all neonates were randomized to maternal milk only or maternal milk with up to 20 supplemental formula feedings. All pups remained with the Dams and were additionally allowed to nurse ad libitum. RESULTS Formula-fed F2NONE pups rarely developed NEC while this disease was common in formula-fed F2MTCDD and F2PTCDD mice. Unexpectedly, 50% of F2MTCDD pups that were not provided supplemental formula also developed NEC. CONCLUSIONS Our studies provide evidence that a history of parental TCDD exposure enhances the risk of NEC in offspring and suggest exposure to environmental immunotoxicants such as TCDD may also contribute to this inflammatory disease in humans.
Collapse
Affiliation(s)
- Shilpa Mokshagundam
- Women's Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Tianbing Ding
- Women's Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jelonia T Rumph
- Department of Immunology, Microbiology and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | | | - Victoria R Stephens
- Women's Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kevin G Osteen
- Women's Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Kaylon L Bruner-Tran
- Women's Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
47
|
Dornelles LV, Procianoy RS, Roesch LFW, Corso AL, Dobbler PT, Mai V, Silveira RC. Meconium microbiota predicts clinical early-onset neonatal sepsis in preterm neonates. J Matern Fetal Neonatal Med 2020; 35:1935-1943. [PMID: 32508165 DOI: 10.1080/14767058.2020.1774870] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: Early-onset neonatal sepsis (EONS) remains one of the leading causes of morbidity and mortality related to premature birth, and its diagnosis remains difficult. Our goal was to evaluate the intestinal microbiota of the first meconium of preterm newborns and ascertain whether it is associated with clinical EONS.Methods: In a controlled, prospective cohort study, samples of the first meconium of premature infants with a gestational age (GA) ≤32 weeks was obtained at Hospital de Clínicas de Porto Alegre and DNA was isolated from the samples. 16S rDNA based microbiota composition of preterm infants with a clinical diagnosis of EONS was compared to that of a control group.Results: 40 (48%) premature infants with clinical diagnosis of EONS and 44 (52%) without EONS were included in the analysis. The most abundant phylum detected in both groups, Proteobacteria, was more prevalent in the sepsis group (p = .034). 14% of variance among bacterial communities (p = .001) correlated with EONS. The genera most strongly associated with EONS were Paenibacillus, Caulobacter, Dialister, Akkermansia, Phenylobacterium, Propionibacterium, Ruminococcus, Bradyrhizobium, and Alloprevotella. A single genus, Flavobacterium, was most strongly associated with the control group.Conclusion: These findings suggest that the first-meconium microbiota is different in preterm neonates with and without clinical EONS.
Collapse
Affiliation(s)
- Laura V Dornelles
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Programa de Pós Graduação em Saúde da Criança e do Adolescente, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Renato S Procianoy
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Programa de Pós Graduação em Saúde da Criança e do Adolescente, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiz F W Roesch
- Centro Interdisciplinar de Pesquisas em Biotecnologia - CIP-Biotec, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, Brazil
| | - Andréa L Corso
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Programa de Pós Graduação em Saúde da Criança e do Adolescente, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Centro Interdisciplinar de Pesquisas em Biotecnologia - CIP-Biotec, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, Brazil
| | - Priscila Thiago Dobbler
- Centro Interdisciplinar de Pesquisas em Biotecnologia - CIP-Biotec, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, Brazil
| | - Volker Mai
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Rita C Silveira
- Serviço de Neonatologia do Hospital de Clínicas de Porto Alegre, Programa de Pós Graduação em Saúde da Criança e do Adolescente, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
48
|
Chen Y, Brook TC, Soe CZ, O'Neill I, Alcon-Giner C, Leelastwattanagul O, Phillips S, Caim S, Clarke P, Hall LJ, Hoyles L. Preterm infants harbour diverse Klebsiella populations, including atypical species that encode and produce an array of antimicrobial resistance- and virulence-associated factors. Microb Genom 2020; 6:e000377. [PMID: 32436839 PMCID: PMC7371107 DOI: 10.1099/mgen.0.000377] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/26/2020] [Indexed: 01/08/2023] Open
Abstract
Klebsiella spp. are frequently enriched in the gut microbiota of preterm neonates, and overgrowth is associated with necrotizing enterocolitis (NEC), nosocomial infections and late-onset sepsis. Little is known about the genomic and phenotypic characteristics of preterm-associated Klebsiella, as previous studies have focused on the recovery of antimicrobial-resistant isolates or culture-independent molecular analyses. The aim of this study was to better characterize preterm-associated Klebsiella populations using phenotypic and genotypic approaches. Faecal samples from a UK cohort of healthy and sick preterm neonates (n=109) were screened on MacConkey agar to isolate lactose-positive Enterobacteriaceae. Whole-genome sequences were generated for Klebsiella spp., and virulence and antimicrobial resistance genes identified. Antibiotic susceptibility profiling and in vitro macrophage and iron assays were undertaken for the Klebsiella strains. Metapangenome analyses with a manually curated genome dataset were undertaken to examine the diversity of Klebsiella oxytoca and related bacteria in a publicly available shotgun metagenome dataset. Approximately one-tenth of faecal samples harboured Klebsiella spp. (Klebsiella pneumoniae, 7.3 %; Klebsiella quasipneumoniae, 0.9 %; Klebsiella grimontii, 2.8 %; Klebsiella michiganensis, 1.8 %). Isolates recovered from NEC- and sepsis-affected infants and those showing no signs of clinical infection (i.e. 'healthy') encoded multiple β-lactamases. No difference was observed between isolates recovered from healthy and sick infants with respect to in vitro siderophore production (all encoded enterobactin in their genomes). All K. pneumoniae, K. quasipneumoniae, K. grimontii and K. michiganensis faecal isolates tested were able to reside and persist in macrophages, indicating their immune evasion abilities. Metapangenome analyses of published metagenomic data confirmed our findings regarding the presence of K. michiganensis in the preterm gut. There is little difference in the phenotypic and genomic characteristics of Klebsiella isolates recovered from healthy and sick infants. Identification of β-lactamases in all isolates may prove problematic when defining treatment regimens for NEC or sepsis, and suggests that healthy preterm infants contribute to the resistome. Refined analyses with curated sequence databases are required when studying closely related species present in metagenomic data.
Collapse
Affiliation(s)
- Yuhao Chen
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Thomas C. Brook
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, London, UK
| | - Cho Zin Soe
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Ian O'Neill
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Cristina Alcon-Giner
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Onnicha Leelastwattanagul
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi (Bang Khun Thian Campus), Bangkok, Thailand
| | - Sarah Phillips
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Shabhonam Caim
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Paul Clarke
- Neonatal Intensive Care Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Lindsay J. Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Lesley Hoyles
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
49
|
Bacterial colonization reprograms the neonatal gut metabolome. Nat Microbiol 2020; 5:838-847. [PMID: 32284564 PMCID: PMC8052915 DOI: 10.1038/s41564-020-0694-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022]
Abstract
Initial microbial colonization and later succession in the gut of human infants are linked to health and disease later in life. The timing of the appearance of the first gut microbiome, and the consequences for the early life metabolome, are just starting to be defined. Here, we evaluated the gut microbiome, proteome and metabolome in 88 African-American newborns using faecal samples collected in the first few days of life. Gut bacteria became detectable using molecular methods by 16 h after birth. Detailed analysis of the three most common species, Escherichia coli, Enterococcus faecalis and Bacteroides vulgatus, did not suggest a genomic signature for neonatal gut colonization. The appearance of bacteria was associated with reduced abundance of approximately 50 human proteins, decreased levels of free amino acids and an increase in products of bacterial fermentation, including acetate and succinate. Using flux balance modelling and in vitro experiments, we provide evidence that fermentation of amino acids provides a mechanism for the initial growth of E. coli, the most common early colonizer, under anaerobic conditions. These results provide a deep characterization of the first microbes in the human gut and show how the biochemical environment is altered by their appearance.
Collapse
|
50
|
Hooven TA, Lin AYC, Salleb-Aouissi A. Multiple Instance Learning for Predicting Necrotizing Enterocolitis in Premature Infants Using Microbiome Data. PROCEEDINGS OF THE ACM CONFERENCE ON HEALTH, INFERENCE, AND LEARNING 2020; 2020:99-109. [PMID: 34318306 PMCID: PMC8313028 DOI: 10.1145/3368555.3384466] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening intestinal disease that primarily affects preterm infants during their first weeks after birth. Mortality rates associated with NEC are 15-30%, and surviving infants are susceptible to multiple serious, long-term complications. The disease is sporadic and, with currently available tools, unpredictable. We are creating an early warning system that uses stool microbiome features, combined with clinical and demographic information, to identify infants at high risk of developing NEC. Our approach uses a multiple instance learning, neural network-based system that could be used to generate daily or weekly NEC predictions for premature infants. The approach was selected to effectively utilize sparse and weakly annotated datasets characteristic of stool microbiome analysis. Here we describe initial validation of our system, using clinical and microbiome data from a nested case-control study of 161 preterm infants. We show receiver-operator curve areas above 0.9, with 75% of dominant predictive samples for NEC-affected infants identified at least 24 hours prior to disease onset. Our results pave the way for development of a real-time early warning system for NEC using a limited set of basic clinical and demographic details combined with stool microbiome data.
Collapse
|