Neonatal Microbiome, Intestinal Inflammation, and Necrotizing Enterocolitis (NEC)

A special issue of Microorganisms (ISSN 2076-2607). This special issue belongs to the section "Medical Microbiology".

Deadline for manuscript submissions: closed (31 July 2023) | Viewed by 14165

Special Issue Editors


E-Mail Website
Guest Editor
Department of Pediatrics, Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
Interests: neonatal necrotizing enterocolitis (NEC); glycosaminoglycans; intestinal stem cells; hypoxia; human milk bioactive factors

E-Mail Website
Guest Editor
Indiana University School of Medicine, Indianapolis, IN, USA
Interests: necrotizing enterocolitis; mesenchymal stem cells; hypoxia; vascular endothelial growth factor; ischemia and reperfusion; enteral feeds; short bowel syndrome

Special Issue Information

Dear Colleagues,

NEC is a leading cause of preterm infant morbidity and mortality, and the most common gastrointestinal emergency in the neonatal intensive care unit (NICU). While treatment options are currently limited to supportive therapy, the latest research has implicated a dysbiotic microbiome in NEC pathogenesis. In this Special Issue of Microorganisms, we invite you to send contributions concerning the latest information on neonatal microbiome development, particularly in the preterm infant, host characteristics influencing microbiome composition, and microbiome effects on the preterm immune system, intestinal epithelium, and barrier function. In addition, the role of specific microbiota, whether original colonizers or targeted probiotic supplements, in mitigating NEC risk will be included. This issue seeks to provide a comprehensive composite of the advances in host microbiome development, modifiable influences on microbiome composition, such as mode of feeding, and the potential for targeted pre- and probiotics in modulating or preventing NEC.

In this Special Issue, original research articles and reviews are welcome.

We look forward to receiving your contributions.

Dr. Kathryn Y. Burge
Dr. Troy A. Markel
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Microorganisms is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • necrotizing enterocolitis
  • infant microbiome
  • mode of feeding
  • human milk
  • probiotics
  • prebiotics
  • prematurity
  • host immune response
  • inflammation
  • antibiotics

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

2 pages, 168 KiB  
Editorial
Neonatal Microbiome, Intestinal Inflammation, and Necrotizing Enterocolitis
by Kathryn Y. Burge and Troy A. Markel
Microorganisms 2022, 10(7), 1382; https://doi.org/10.3390/microorganisms10071382 - 9 Jul 2022
Viewed by 1157
Abstract
Necrotizing enterocolitis (NEC), the most common gastrointestinal emergency in the neonatal intensive care unit (NICU), is a leading cause of preterm infant morbidity and mortality [...] Full article

Research

Jump to: Editorial, Review

14 pages, 1765 KiB  
Article
Occurrence of Neonatal Necrotizing Enterocolitis in Premature Neonates and Gut Microbiota: A Case–Control Prospective Multicenter Study
by Julio Aires, Zehra Esra Ilhan, Lancelot Nicolas, Laurent Ferraris, Johanne Delannoy, Maxime Bredel, Anne Chauvire-Drouard, Frédéric Barbut, Jean-Christophe Rozé, Patricia Lepage, Marie-José Butel and ClosNEC Study Group
Microorganisms 2023, 11(10), 2457; https://doi.org/10.3390/microorganisms11102457 - 29 Sep 2023
Viewed by 1083
Abstract
Background: Necrotizing enterocolitis (NEC) is still one of the leading causes of neonatal death. The present study reports the data from a French case–control prospective multicenter study. Methods: A total of 146 preterm neonates (PNs) with or without NEC were included. Bacterial 16S [...] Read more.
Background: Necrotizing enterocolitis (NEC) is still one of the leading causes of neonatal death. The present study reports the data from a French case–control prospective multicenter study. Methods: A total of 146 preterm neonates (PNs) with or without NEC were included. Bacterial 16S rRNA gene sequencing was performed on stool samples (n = 103). Specific culture media were used to isolate Escherichia coli, Clostridium butyricum, and Clostridium neonatale, and strains were phenotypically characterized. Results: The gut microbiota of PNs was dominated by Firmicutes and Proteobacteria, and five enterotypes were identified. The microbiota composition was similar between NEC cases and PN controls. However, differences were observed in the relative abundance of Lactobacillus genus, which was significantly lower in the NEC group, whereas that of the Clostridium cluster III was significantly higher (p < 0.05). Within enterotypes, several phylotypes were significantly more abundant in NEC cases (p < 0.05). Regarding perinatal factors, a statistical association was found between the gut microbiota and cesarean delivery and antifungal therapy. In NEC cases and PN controls, the carriage rates and virulence genes of uropathogenic E. coli were equivalent based on culture. No correlation was found between E. coli, C. butyricum, and C. neonatale carriages, beta-lactam resistance, and antibiotic treatment. Conclusions: At disease onset, our data support a microbiota dysbiosis between NEC and control infants at the genus level. In addition, it provides valuable information on bacterial antimicrobial susceptibility. Full article
Show Figures

Figure 1

17 pages, 3769 KiB  
Article
The Detrimental Effects of Peripartum Antibiotics on Gut Proliferation and Formula Feeding Injury in Neonatal Mice Are Alleviated with Lactobacillus rhamnosus GG
by Alain Cuna, Marianne Nsumu, Heather L. Menden, Susana Chavez-Bueno and Venkatesh Sampath
Microorganisms 2023, 11(6), 1482; https://doi.org/10.3390/microorganisms11061482 - 1 Jun 2023
Viewed by 1459
Abstract
Peripartum antibiotics can negatively impact the developing gut microbiome and are associated with necrotizing enterocolitis (NEC). The mechanisms by which peripartum antibiotics increase the risk of NEC and strategies that can help mitigate this risk remain poorly understood. In this study, we determined [...] Read more.
Peripartum antibiotics can negatively impact the developing gut microbiome and are associated with necrotizing enterocolitis (NEC). The mechanisms by which peripartum antibiotics increase the risk of NEC and strategies that can help mitigate this risk remain poorly understood. In this study, we determined mechanisms by which peripartum antibiotics increase neonatal gut injury and evaluated whether probiotics protect against gut injury potentiated by peripartum antibiotics. To accomplish this objective, we administered broad-spectrum antibiotics or sterile water to pregnant C57BL6 mice and induced neonatal gut injury to their pups with formula feeding. We found that pups exposed to antibiotics had reduced villus height, crypt depth, and intestinal olfactomedin 4 and proliferating cell nuclear antigen compared to the controls, indicating that peripartum antibiotics impaired intestinal proliferation. When formula feeding was used to induce NEC-like injury, more severe intestinal injury and apoptosis were observed in the pups exposed to antibiotics compared to the controls. Supplementation with the probiotic Lactobacillus rhamnosus GG (LGG) reduced the severity of formula-induced gut injury potentiated by antibiotics. Increased intestinal proliferating cell nuclear antigen and activation of the Gpr81-Wnt pathway were noted in the pups supplemented with LGG, suggesting partial restoration of intestinal proliferation by probiotics. We conclude that peripartum antibiotics potentiate neonatal gut injury by inhibiting intestinal proliferation. LGG supplementation decreases gut injury by activating the Gpr81-Wnt pathway and restoring intestinal proliferation impaired by peripartum antibiotics. Our results suggest that postnatal probiotics may be effective in mitigating the increased risk of NEC associated with peripartum antibiotic exposure in preterm infants. Full article
Show Figures

Figure 1

24 pages, 12858 KiB  
Article
Microbiota from Preterm Infants Who Develop Necrotizing Enterocolitis Drives the Neurodevelopment Impairment in a Humanized Mouse Model
by Jing Lu, Alexander Drobyshevsky, Lei Lu, Yueyue Yu, Michael S. Caplan and Erika C. Claud
Microorganisms 2023, 11(5), 1131; https://doi.org/10.3390/microorganisms11051131 - 26 Apr 2023
Cited by 5 | Viewed by 2118
Abstract
Necrotizing enterocolitis (NEC) is the leading basis for gastrointestinal morbidity and poses a significant risk for neurodevelopmental impairment (NDI) in preterm infants. Aberrant bacterial colonization preceding NEC contributes to the pathogenesis of NEC, and we have demonstrated that immature microbiota in preterm infants [...] Read more.
Necrotizing enterocolitis (NEC) is the leading basis for gastrointestinal morbidity and poses a significant risk for neurodevelopmental impairment (NDI) in preterm infants. Aberrant bacterial colonization preceding NEC contributes to the pathogenesis of NEC, and we have demonstrated that immature microbiota in preterm infants negatively impacts neurodevelopment and neurological outcomes. In this study, we tested the hypothesis that microbial communities before the onset of NEC drive NDI. Using our humanized gnotobiotic model in which human infant microbial samples were gavaged to pregnant germ-free C57BL/6J dams, we compared the effects of the microbiota from preterm infants who went on to develop NEC (MNEC) to the microbiota from healthy term infants (MTERM) on brain development and neurological outcomes in offspring mice. Immunohistochemical studies demonstrated that MNEC mice had significantly decreased occludin and ZO-1 expression compared to MTERM mice and increased ileal inflammation marked by the increased nuclear phospho-p65 of NFκB expression, revealing that microbial communities from patients who developed NEC had a negative effect on ileal barrier development and homeostasis. In open field and elevated plus maze tests, MNEC mice had worse mobility and were more anxious than MTERM mice. In cued fear conditioning tests, MNEC mice had worse contextual memory than MTERM mice. MRI revealed that MNEC mice had decreased myelination in major white and grey matter structures and lower fractional anisotropy values in white matter areas, demonstrating delayed brain maturation and organization. MNEC also altered the metabolic profiles, especially carnitine, phosphocholine, and bile acid analogs in the brain. Our data demonstrated numerous significant differences in gut maturity, brain metabolic profiles, brain maturation and organization, and behaviors between MTERM and MNEC mice. Our study suggests that the microbiome before the onset of NEC has negative impacts on brain development and neurological outcomes and can be a prospective target to improve long-term developmental outcomes. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research

12 pages, 311 KiB  
Review
The Impact of Stress, Microbial Dysbiosis, and Inflammation on Necrotizing Enterocolitis
by Venkata Yeramilli, Riadh Cheddadi, Heather Benjamin and Colin Martin
Microorganisms 2023, 11(9), 2206; https://doi.org/10.3390/microorganisms11092206 - 31 Aug 2023
Cited by 1 | Viewed by 1032
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 [...] Read more.
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. Full article
25 pages, 1144 KiB  
Review
Volatile Organic Compound Assessment as a Screening Tool for Early Detection of Gastrointestinal Diseases
by Costa Dalis, Fikir M. Mesfin, Krishna Manohar, Jianyun Liu, W. Christopher Shelley, John P. Brokaw and Troy A. Markel
Microorganisms 2023, 11(7), 1822; https://doi.org/10.3390/microorganisms11071822 - 17 Jul 2023
Cited by 3 | Viewed by 1877
Abstract
Gastrointestinal (GI) diseases have a high prevalence throughout the United States. Screening and diagnostic modalities are often expensive and invasive, and therefore, people do not utilize them effectively. Lack of proper screening and diagnostic assessment may lead to delays in diagnosis, more advanced [...] Read more.
Gastrointestinal (GI) diseases have a high prevalence throughout the United States. Screening and diagnostic modalities are often expensive and invasive, and therefore, people do not utilize them effectively. Lack of proper screening and diagnostic assessment may lead to delays in diagnosis, more advanced disease at the time of diagnosis, and higher morbidity and mortality rates. Research on the intestinal microbiome has demonstrated that dysbiosis, or unfavorable alteration of organismal composition, precedes the onset of clinical symptoms for various GI diseases. GI disease diagnostic research has led to a shift towards non-invasive methods for GI screening, including chemical-detection tests that measure changes in volatile organic compounds (VOCs), which are the byproducts of bacterial metabolism that result in the distinct smell of stool. Many of these tools are expensive, immobile benchtop instruments that require highly trained individuals to interpret the results. These attributes make them difficult to implement in clinical settings. Alternatively, electronic noses (E-noses) are relatively cheaper, handheld devices that utilize multi-sensor arrays and pattern recognition technology to analyze VOCs. The purpose of this review is to (1) highlight how dysbiosis impacts intestinal diseases and how VOC metabolites can be utilized to detect alterations in the microbiome, (2) summarize the available VOC analytical platforms that can be used to detect aberrancies in intestinal health, (3) define the current technological advancements and limitations of E-nose technology, and finally, (4) review the literature surrounding several intestinal diseases in which headspace VOCs can be used to detect or predict disease. Full article
Show Figures

Figure 1

15 pages, 1119 KiB  
Review
Development of the Neonatal Intestinal Barrier, Microbiome, and Susceptibility to NEC
by Alena Golubkova and Catherine J. Hunter
Microorganisms 2023, 11(5), 1247; https://doi.org/10.3390/microorganisms11051247 - 9 May 2023
Cited by 5 | Viewed by 2083
Abstract
The function of the intestinal barrier is partially dependent on host maturity and the colonization patterns of the microbiome to which it is exposed. Premature birth and stressors of neonatal intensive care unit (NICU)-related support (e.g., antibiotics, steroids, etc.) can alter the host [...] Read more.
The function of the intestinal barrier is partially dependent on host maturity and the colonization patterns of the microbiome to which it is exposed. Premature birth and stressors of neonatal intensive care unit (NICU)-related support (e.g., antibiotics, steroids, etc.) can alter the host internal environment resulting in changes in the intestinal barrier. Pathogenic microbial proliferation and breach of the immature intestinal barrier are proposed to be crucial steps in the development of neonatal diseases such as necrotizing enterocolitis. This article will review the current literature on the intestinal barrier in the neonatal gut, the consequences of microbiome development for this defense system, and how prematurity can influence neonatal susceptibility to gastrointestinal infection. Full article
Show Figures

Figure 1

21 pages, 458 KiB  
Review
The Nonbacterial Microbiome: Fungal and Viral Contributions to the Preterm Infant Gut in Health and Disease
by Adam Wilson, Brett Bogie, Hala Chaaban and Kathryn Burge
Microorganisms 2023, 11(4), 909; https://doi.org/10.3390/microorganisms11040909 - 31 Mar 2023
Cited by 2 | Viewed by 2361
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 [...] Read more.
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. Full article
Back to TopTop