Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 410
Filter
1.
Cell Host Microbe ; 32(10): 1805-1821.e10, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39293437

ABSTRACT

Microbiota and feeding modes influence the susceptibility of premature newborns to necrotizing enterocolitis (NEC) through mechanisms that remain unknown. Here, we show that microbiota colonization facilitated by breastmilk feeding promotes NOD-like receptor family CARD domain containing 5 (Nlrc5) gene expression in mouse intestinal epithelial cells (IECs). Notably, inducible knockout of the Nlrc5 gene in IECs predisposes neonatal mice to NEC-like injury in the small intestine upon viral inflammation in an NK1.1+ cell-dependent manner. By contrast, formula feeding enhances neonatal gut colonization with environment-derived tilivalline-producing Klebsiella spp. Remarkably, tilivalline disrupts microbiota-activated STAT1 signaling that controls Nlrc5 gene expression in IECs through a PPAR-γ-mediated mechanism. Consequently, this dysregulation hinders the resistance of neonatal intestinal epithelium to self-NK1.1+ cell cytotoxicity upon virus infection/colonization, promoting NEC development. Together, we discover the underappreciated role of intestinal microbiota colonization in shaping a disease tolerance program to viral inflammation and elucidate the mechanisms impacting NEC development in neonates.


Subject(s)
Animals, Newborn , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Intestinal Mucosa , STAT1 Transcription Factor , Animals , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/virology , STAT1 Transcription Factor/metabolism , Mice , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Knockout , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Signal Transduction , Epithelial Cells/microbiology , Epithelial Cells/virology , Epithelial Cells/immunology , Humans , Mice, Inbred C57BL
2.
Cell Host Microbe ; 32(10): 1822-1837.e5, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39197454

ABSTRACT

Aberrant preterm infant gut microbiota assembly predisposes to early-life disorders and persistent health problems. Here, we characterize gut microbiome dynamics over the first 3 months of life in 236 preterm infants hospitalized in three neonatal intensive care units using shotgun metagenomics of 2,512 stools and metatranscriptomics of 1,381 stools. Strain tracking, taxonomic and functional profiling, and comprehensive clinical metadata identify Enterobacteriaceae, enterococci, and staphylococci as primarily exploiting available niches to populate the gut microbiome. Clostridioides difficile lineages persist between individuals in single centers, and Staphylococcus epidermidis lineages persist within and, unexpectedly, between centers. Collectively, antibiotic and non-antibiotic medications influence gut microbiome composition to greater extents than maternal or baseline variables. Finally, we identify a persistent low-diversity gut microbiome in neonates who develop necrotizing enterocolitis after day of life 40. Overall, we comprehensively describe gut microbiome dynamics in response to medical interventions in preterm, hospitalized neonates.


Subject(s)
Anti-Bacterial Agents , Enterocolitis, Necrotizing , Feces , Gastrointestinal Microbiome , Infant, Premature , Metagenomics , Humans , Infant, Newborn , Feces/microbiology , Enterocolitis, Necrotizing/microbiology , Female , Anti-Bacterial Agents/pharmacology , Male , Clostridioides difficile/genetics , Intensive Care Units, Neonatal , Infant , Hospitalization , Enterobacteriaceae/genetics , Enterococcus/genetics , Staphylococcus epidermidis , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification
3.
J Hosp Infect ; 152: 21-27, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094736

ABSTRACT

BACKGROUND: Necrotizing enterocolitis is the most severe life-threatening acquired gastrointestinal disorder among preterm neonates. We describe here an outbreak of Clostridium butyricum-related necrotizing enterocolitis in preterm neonates that occurred in three different neonatal centres, in southeast France. METHODS: We defined a confirmed case of C. butyricum-related necrotizing enterocolitis in preterm neonates by the presence of clinical signs according to modified Bell criteria and C. butyricum identified from stool samples using real-time polymerase chain reaction or culture. A phylogenetic analysis of the isolated strains by whole-genome sequencing was also performed. RESULTS: Between 5th and 27th January 2022, we identified 10 confirmed cases of C. butyricum-related necrotizing enterocolitis, including five from Neonatal Centre 1, four from Neonatal Centre 2, and one from Neonatal Centre 3. The attack rate of necrotizing enterocolitis in Neonatal Centre 1 was 7.1% (5/70). The positivity rate of C. butyricum detected from stool samples was higher during the outbreak period (37/276; 13.4%) than outside this period (7/369; 1.9%), while systematic screening was maintained (P<0.001). Phylogenetic analysis showed a clonality between strains inside four clusters. Two clusters included neonates hospitalized in different neonatal centres, suggesting the transmission of C. butyricum strains during the transfer of neonates between neonatal centres. CONCLUSIONS: This outbreak of C. butyricum-related necrotizing enterocolitis confirms a cross-transmission between preterm neonates, including twin or triplet siblings, and involving necrotizing enterocolitis cases together with asymptomatic carriers. After three months of follow-up, no further cases were identified following the implementation of contact precautions with sporicidal agents.


Subject(s)
Clostridium Infections , Clostridium butyricum , Disease Outbreaks , Enterocolitis, Necrotizing , Feces , Infant, Premature , Phylogeny , Whole Genome Sequencing , Humans , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/epidemiology , Infant, Newborn , France/epidemiology , Clostridium butyricum/genetics , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Clostridium Infections/transmission , Male , Female , Feces/microbiology , Disease Transmission, Infectious , Cross Infection/epidemiology , Cross Infection/microbiology , Cross Infection/transmission
4.
Cell Rep Med ; 5(9): 101708, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39216480

ABSTRACT

Necrotizing enterocolitis (NEC) is a severe intestinal disease of very preterm infants with mother's own milk (MOM) providing protection, but the contribution of the MOM microbiota to NEC risk has not been explored. Here, we analyze MOM of 110 preterm infants (48 NEC, 62 control) in a cross-sectional study. Breast milk contains viable bacteria, but there is no significant difference in MOM microbiota between NEC and controls. Integrative analysis between MOM microbiota, human milk oligosaccharides (HMOs), and the infant gut microbiota shows positive correlations only between Acinetobacter in the infant gut and Acinetobacter and Staphylococcus in MOM. This study suggests that NEC protection from MOM is not modulated through the MOM microbiota. Thus, "'restoring" the MOM microbiota in donor human milk is unlikely to reduce NEC, and emphasis should instead focus on increasing fresh maternal human milk intake and researching different therapies for NEC prevention.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Premature , Milk, Human , Oligosaccharides , Humans , Milk, Human/microbiology , Milk, Human/chemistry , Enterocolitis, Necrotizing/microbiology , Oligosaccharides/metabolism , Infant, Newborn , Female , Male , Cross-Sectional Studies
5.
Adv Exp Med Biol ; 1449: 43-57, 2024.
Article in English | MEDLINE | ID: mdl-39060730

ABSTRACT

Intestinal bacteria, also known as gut microbiota, are a rich ecology of microorganisms found in the human digestive tract. Extensive study has highlighted their critical relevance in preserving human health. New research has revealed that bacterial viability is not invariably necessary to induce health benefits. Postbiotics (defined soluble substances produced as a byproduct of the metabolic processes of living microbes) have thus emerged as an important topic of research. They contribute to shaping the gut microbiota, exert immune-modulation activity, and improve the integrity of the gut barrier.Alterations in preterm gut colonization associated with intestinal barrier immaturity and the increased reactivity of the intestinal mucosa to colonizing bacteria have been implicated in the pathogenesis of necrotizing enterocolitis. Postbiotics have shown promising outcomes in reducing the risk of developing NEC, lowering inflammation, encouraging the development of good bacteria, and strengthening the intestinal barrier. This is an important advancement in newborn care and highlights the potential of postbiotics to avoid severe intestinal disorders.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/immunology , Humans , Infant, Newborn , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Probiotics/therapeutic use , Animals , Infant, Premature , Intestines/microbiology
6.
Adv Exp Med Biol ; 1449: 29-41, 2024.
Article in English | MEDLINE | ID: mdl-39060729

ABSTRACT

Preterm infants differ significantly from their term infant counterparts regarding bacterial colonization patterns related to maternal microbiota diversity, mode of delivery, feeding type, antibiotic exposure, and the environmental influences related to prolonged hospitalization in the neonatal intensive care unit (NICU). Necrotizing enterocolitis (NEC), a multifactorial intestinal disorder characterized by ischemic bowel disease, disproportionately impacts preterm infants and has a high disease burden. Recent studies in the basic, translational, and clinical scientific literature have advanced knowledge into this complex disease process. Despite the explosion of research into NEC, however, there is a still a great deal unknown about this devastating illness. Additionally, the disease morbidity and mortality for NEC remain high despite advances in therapy options. This chapter reviews the current literature into the preterm infant microbiome, pathogenesis of NEC, potential targets for altering preterm microbiome, influence of microbiome on other organ systems, long-term implications of microbiome dysbiosis, and future directions of study.


Subject(s)
Dysbiosis , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Premature , Humans , Enterocolitis, Necrotizing/microbiology , Infant, Newborn , Dysbiosis/microbiology
7.
Gut Microbes ; 16(1): 2379566, 2024.
Article in English | MEDLINE | ID: mdl-39013030

ABSTRACT

Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants with no specific treatments available. We aimed to identify the molecular mechanisms underlying NEC and investigate the therapeutic effects of Bacteroides fragilis on NEC. Clinical samples of infant feces, bile acid-targeted metabolomics, pathological staining, bioinformatics analysis, NEC rat model, and co-immunoprecipitation were used to explore the pathogenesis of NEC. Taxonomic characterization of the bile salt hydrolase (bsh) gene, enzyme activity assays, 16S rRNA sequencing, and organoids were used to explore the therapeutic effects of B. fragilis on NEC-related intestinal damage. Clinical samples, NEC rat models, and in vitro experiments revealed that total bile acid increased in the blood but decreased in feces. Moreover, the levels of FXR and other bile acid metabolism-related genes were abnormal, resulting in disordered bile acid metabolism in NEC. Taurochenodeoxycholic acid accelerated NEC pathogenesis and taurodeoxycholate alleviated NEC. B. fragilis displayed bsh genes and enzyme activity and alleviated intestinal damage by restoring gut microbiota dysbiosis and bile acid metabolism abnormalities by inhibiting the FXR-NLRP3 signaling pathway. Our results provide valuable insights into the therapeutic role of B. fragilis in NEC. Administering B. fragilis may substantially alleviate intestinal damage in NEC.


Subject(s)
Amidohydrolases , Bacteroides fragilis , Bile Acids and Salts , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Cytoplasmic and Nuclear , Signal Transduction , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/drug therapy , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Bacteroides fragilis/metabolism , Bacteroides fragilis/genetics , Signal Transduction/drug effects , Bile Acids and Salts/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Gastrointestinal Microbiome/drug effects , Amidohydrolases/metabolism , Amidohydrolases/genetics , Humans , Rats, Sprague-Dawley , Infant, Newborn , Disease Models, Animal , Male , Female , Probiotics/administration & dosage , Probiotics/pharmacology , Infant, Premature , Dysbiosis/microbiology
8.
Kaohsiung J Med Sci ; 40(9): 780-788, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39073226

ABSTRACT

The gut microbiota undergoes substantial development from birth, and its development in the initial years of life has a potentially lifelong effect on the health of the individual. However, various factors can disrupt the development of the gut microbiota, leading to a condition known as dysbiosis, particularly in preterm infants. Current studies involving adults have suggested that the gut microbiota not only influences the gut but also has multidimensional effects on remote organs; these pathways are often referred to as the gut-organ axis. Imbalance of the gut microbiota may lead to the development of multiple diseases. Recent studies have revealed that gut dysbiosis in preterm infants may cause several acute morbidities-such as necrotizing enterocolitis, late-onset sepsis, bronchopulmonary dysplasia, and retinopathy of prematurity-and it may also influence long-term outcomes including neurodevelopment and somatic growth. This review mainly presents the existing evidence regarding the relationships between the gut microbiota and these morbidities in preterm infants and explores the role of the gut-organ axis in these morbidities. This paper thus offers insights into the future perspectives on microbiota interventions for promoting the health of preterm infants.


Subject(s)
Dysbiosis , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Premature , Humans , Gastrointestinal Microbiome/physiology , Infant, Newborn , Enterocolitis, Necrotizing/microbiology , Dysbiosis/microbiology , Bronchopulmonary Dysplasia/microbiology , Retinopathy of Prematurity/microbiology , Sepsis/microbiology , Morbidity
9.
Front Cell Infect Microbiol ; 14: 1391929, 2024.
Article in English | MEDLINE | ID: mdl-38903936

ABSTRACT

Objective: To assess the effects of COVID-19 pandemic on the epidemiology of neonatal sepsis and the antibiotic resistance profiles of pathogens involved. Methods: This retrospective cohort study analyzed infants diagnosed with culture-proven sepsis at the neonatal department of a tertiary children's hospital in East China from January 2016 to December 2022. We compared the clinical and microbiological characteristics of neonatal sepsis cases between the pre-pandemic Phase I (2016-2019) and during the COVID-19 pandemic Phase II (2020-2022). Results: A total of 507 infants with 525 sepsis episodes were included, with 343 episodes in Phase I and 182 in Phase II. The incidence of early-onset sepsis (EOS) was significantly lower during Phase II (p < 0.05). Infants in Phase II had lower gestational ages and birth weights compared to Phase I. Clinical signs such as mottled skin, severe anemia, thrombocytopenia were more prevalent in Phase II, alongside a higher incidence of complications. Notably, necrotizing enterocolitis (NEC) (p < 0.05) and meningitis (p < 0.1) occurred more frequently during Phase II. Escherichia coli (E. coli) and Klebsiella pneumoniae (K. pneumoniae) were the predominant pathogens isolated from infants of death and cases with complications. A significant decrease in the proportion of K. pneumoniae was observed in Phase II, alongside increased antibiotic resistance in both E. coli and K. pneumoniae. The period of the COVID-19 pandemic (Phase II) was identified as an independent risk factor for complications in infants with neonatal sepsis. Conclusion: COVID-19 pandemic response measures correlated with a decrease in EOS and an increase in neonatal sepsis complications and antibiotic resistance.


Subject(s)
COVID-19 , Neonatal Sepsis , SARS-CoV-2 , Humans , COVID-19/epidemiology , Infant, Newborn , Retrospective Studies , Female , Neonatal Sepsis/epidemiology , Neonatal Sepsis/microbiology , Male , China/epidemiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Incidence , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Enterocolitis, Necrotizing/epidemiology , Enterocolitis, Necrotizing/microbiology , Sepsis/epidemiology , Sepsis/microbiology , Gestational Age , Pandemics , Escherichia coli/isolation & purification , Escherichia coli/drug effects , Drug Resistance, Bacterial
10.
Gut Microbes ; 16(1): 2352175, 2024.
Article in English | MEDLINE | ID: mdl-38743045

ABSTRACT

The role of gut microbiome in health, a century-old concept, has been on the center stage of medical research recently. While different body sites, disease conditions, and populations have been targeted, neonatal and early infancy appear to be the most suitable period for such interventions. It is intriguing to note that, unlike traditional use in diarrhea and maintenance of gastrointestinal health, microbiome-mediating therapies have now addressed the most serious medical conditions in young infants such as necrotizing enterocolitis and neonatal sepsis. Unfortunately, almost all new endeavors in this space have been carried out in the Western world leaving behind millions of neonates that can benefit from such manipulations while serving as a large resource for further learning. In this review, an attempt has been made to quantify the global burden of neonatal morbidity and mortality, examples presented on interventions that have failed as a result of drawing from studies conducted in the West, and a case made for manipulating the neonatal gut microbiome to address the biggest killers in early life. A brief comparative analysis has been made to demonstrate the differences in the gut microbiota of North and South and a large clinical trial of synbiotics conducted by our group in a South Asian setting has been presented. Although challenging, the value of conducting such global health research is introduced with an intent to invite medical scientists to engage in well-planned, scientifically robust research endeavors. This can bring about innovation while saving and serving the most vulnerable citizens now and protecting them from the negative health consequences in the later part of their lives, ultimately shaping a resilient and equitable world as pledged by 193 United Nations member countries in 2015.


Subject(s)
Gastrointestinal Microbiome , Global Health , Humans , Infant, Newborn , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/prevention & control , Infant , Synbiotics/administration & dosage , Neonatal Sepsis/microbiology , Neonatal Sepsis/prevention & control
11.
J Neonatal Perinatal Med ; 17(2): 265-268, 2024.
Article in English | MEDLINE | ID: mdl-38669486

ABSTRACT

 Streptococcus gallolyticus subspecies pasteurianus is a subtype of Streptococcus bovis (S. bovis) that has become increasingly recognized as a sepsis-causing pathogen in neonates. It is well documented that S. bovis species have a predilection to both cardiac and gastrointestinal tissue, and in adult populations, isolating these organisms in the bloodstream often triggers further evaluation for co-morbid complications such as colon cancer or endocarditis. However, no such guidance currently exists in neonatal literature. We present a case of a preterm infant with S. gallolyticus subsp. pasteurianus bacteremia presenting as necrotizing enterocolitis (NEC) not previously described in the literature. Furthermore, through a complete diagnostic evaluation, including an echocardiogram, our patient was found to have the rare complication of endocarditis.


Subject(s)
Enterocolitis, Necrotizing , Infant, Premature , Streptococcal Infections , Humans , Infant, Newborn , Anti-Bacterial Agents/therapeutic use , Arteritis/microbiology , Bacteremia/microbiology , Enterocolitis, Necrotizing/microbiology , Infant, Premature, Diseases/microbiology , Streptococcal Infections/complications , Streptococcal Infections/microbiology , Streptococcus gallolyticus subspecies gallolyticus
12.
Front Immunol ; 14: 1261010, 2023.
Article in English | MEDLINE | ID: mdl-37841247

ABSTRACT

Necrotizing enterocolitis (NEC) is an inflammatory intestinal disease that profoundly affects preterm infants. Currently, the pathogenesis of NEC remains controversial, resulting in limited treatment strategies. The preterm infants are thought to be susceptible to gut inflammatory disorders because of their immature immune system. In early life, intestinal macrophages (IMφs), crucial components of innate immunity, demonstrate functional plasticity and diversity in intestinal development, resistance to pathogens, maintenance of the intestinal barrier, and regulation of gut microbiota. When the stimulations of environmental, dietary, and bacterial factors interrupt the homeostatic processes of IMφs, they will lead to intestinal disease, such as NEC. This review focuses on the IMφs related pathogenesis in NEC, discusses the multi-functional roles and relevant molecular mechanisms of IMφs in preterm infants, and explores promising therapeutic application for NEC.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Newborn, Diseases , Infant , Infant, Newborn , Humans , Infant, Premature , Enterocolitis, Necrotizing/microbiology , Macrophages
13.
Georgian Med News ; (340-341): 17-24, 2023.
Article in English | MEDLINE | ID: mdl-37805868

ABSTRACT

A gut-brain axis (GBA) has a long history of conceptual development. Intestinal dysbiosis has now been recognized as a key player in the development of adult neurodevelopmental disorders, obesity, and inflammatory bowel disease. Recent developments in metagenomics suggest those nutrition and gut microbiotas (GM) are important regulators of the gut-brain communication pathways that cause neurodevelopmental and psychiatric problems in adulthood. Intestinal dysbiosis and neurodevelopmental disease outcomes in preterm newborns are being linked by recent research. Recent clinical investigations demonstrate that in critical care units, intestinal dysbiosis occurs before late-onset newborn sepsis and necrotizing enterocolitis. Strong epidemiologic data also shows a connection between necrotizing enterocolitis and extremely low birth weight babies' long-term psychomotor impairments and late-onset neonatal sepsis. The GBA theory suggests that intestinal bacteria may indirectly affect preterm newborns' developing brains. In this review, we emphasize the structure and function of the GBA and discuss how immune-microbial dysfunction in the gut affects the transmission of stress signals to the brain. Preterm babies who are exposed to these signals develop neurologic disorders. Understanding neuronal and humoral communication through the GBA may provide insight into therapeutic and nutritional strategies that may enhance the results of very low-birth-weight babies.


Subject(s)
Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Neonatal Sepsis , Nervous System Diseases , Infant , Adult , Infant, Newborn , Humans , Brain-Gut Axis , Enterocolitis, Necrotizing/microbiology , Dysbiosis , Mental Health , Immunity
14.
ACS Appl Mater Interfaces ; 15(30): 35847-35859, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37480336

ABSTRACT

Colostrum provides bioactive components that are essential for the colonization of microbiota in the infant gut, while preventing infectious diseases such as necrotizing enterocolitis. As colostrum is not always available from the mother, particularly for premature infants, effective and safe substitutes are keenly sought after by neonatologists. The benefits of bioactive factors in colostrum are recognized; however, there have been no accounts of human colostrum being studied during digestion of the lipid components or their self-assembly in gastrointestinal environments. Due to the weaker bile pool in infants than adults, evaluating the lipid composition of human colostrum and linking it to structural self-assembly behavior is important in these settings and thus enabling the formulation of substitutes for colostrum. This study is aimed at the rational design of an appropriate lipid component for a colostrum substitute and determining the ability of this formulation to reduce inflammation in intestinal cells. Gas chromatography was utilized to map lipid composition. The self-assembly of lipid components occurring during digestion of colostrum was monitored using small-angle X-ray scattering for comparison with substitute mixtures containing pure triglyceride lipids based on their abundance in colostrum. The digestion profiles of human colostrum and the substitute mixtures were similar. Subtle differences in lipid self-assembly were evident, with the substitute mixtures exhibiting additional non-lamellar phases, which were not seen for human colostrum. The difference is attributable to the distribution of free fatty acids released during digestion. The biological markers of necrotizing enterocolitis were modulated in cells that were treated with bifidobacteria cultured on colostrum substitute mixtures, compared to those treated with infant formula. These findings provide an insight into a colostrum substitute mixture that resembles human colostrum in terms of composition and structural behavior during digestion and potentially reduces some of the characteristics associated with necrotizing enterocolitis.


Subject(s)
Colostrum , Enterocolitis, Necrotizing , Animals , Pregnancy , Female , Infant, Newborn , Humans , Animals, Newborn , Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/microbiology , Inflammation/prevention & control , Lipids
15.
Semin Pediatr Surg ; 32(3): 151309, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37290338

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disorder in premature infants that causes significant morbidity and mortality. Research efforts into the pathogenesis of NEC have discovered a pivotal role for the gram-negative bacterial receptor, Toll-like receptor 4 (TLR4), in its development. TLR4 is activated by dysbiotic microbes within the intestinal lumen, which leads to an exaggerated inflammatory response within the developing intestine, resulting in mucosal injury. More recently, studies have identified that the impaired intestinal motility that occurs early in NEC has a causative role in disease development, as strategies to enhance intestinal motility can reverse NEC in preclinical models. There has also been broad appreciation that NEC also contributes to significant neuroinflammation, which we have linked to the effects of gut-derived pro-inflammatory molecules and immune cells which activate microglia in the developing brain, resulting in white matter injury. These findings suggest that the management of the intestinal inflammation may secondarily be neuroprotective. Importantly, despite the significant burden of NEC on premature infants, these and other studies have provided a strong rationale for the development of small molecules with the capability of reducing NEC severity in pre-clinical models, thus guiding the development of specific anti-NEC therapies. This review summarizes the roles of TLR4 signaling in the premature gut in the pathogenesis of NEC, and provides insights into optimal clinical management strategies based upon findings from laboratory studies.


Subject(s)
Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Infant , Infant, Newborn , Humans , Intestinal Mucosa/pathology , Toll-Like Receptor 4/therapeutic use , Enterocolitis, Necrotizing/therapy , Enterocolitis, Necrotizing/microbiology , Intestines , Infant, Premature , Infant, Newborn, Diseases/pathology
16.
Semin Pediatr Surg ; 32(3): 151307, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37295299

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.


Subject(s)
Enterocolitis, Necrotizing , Inflammatory Bowel Diseases , Probiotics , Female , Infant, Newborn , Humans , Probiotics/therapeutic use , Prebiotics , Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/microbiology , Milk, Human
17.
Sci Rep ; 13(1): 7893, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37193703

ABSTRACT

Intestinal colonization with Klebsiella has been linked to necrotizing enterocolitis (NEC), but methods of analysis usually failed to discriminate Klebsiella species or strains. A novel ~ 2500-base amplicon (StrainID) that spans the 16S and 23S rRNA genes was used to generate amplicon sequence variant (ASV) fingerprints for Klebsiella oxytoca and Klebsiella pneumoniae species complexes (KoSC and KpSC, respectively) and co-occurring fecal bacterial strains from 10 preterm infants with NEC and 20 matched controls. Complementary approaches were used to identify cytotoxin-producing isolates of KoSC. Klebsiella species colonized most preterm infants, were more prevalent in NEC subjects versus controls, and replaced Escherichia in NEC subjects. Single KoSC or KpSC ASV fingerprinted strains dominated the gut microbiota, suggesting exclusionary Klebsiella competition for luminal resources. Enterococcus faecalis was co-dominant with KoSC but present infrequently with KpSC. Cytotoxin-producing KoSC members were identified in most NEC subjects and were less frequent in controls. Few Klebsiella strains were shared between subjects. We conclude that inter-species Klebsiella competition, within an environment of KoSC and E. faecalis cooperation, appears to be an important factor for the development of NEC. Preterm infants seem to acquire Klebsiella primarily through routes other than patient-to-patient transmission.


Subject(s)
Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Microbiota , Infant , Female , Infant, Newborn , Humans , Infant, Premature , Klebsiella/genetics , Enterocolitis, Necrotizing/microbiology , RNA, Ribosomal, 16S/genetics , Microbiota/genetics , Feces/microbiology
18.
Gut Microbes ; 15(1): 2172666, 2023.
Article in English | MEDLINE | ID: mdl-36801067

ABSTRACT

Bacterial colonization in the gut plays a pivotal role in neonatal necrotizing enterocolitis (NEC) development, but the relationship between bacteria and NEC remains unclear. In this study, we aimed to elucidate whether bacterial butyrate end-fermentation metabolites participate in the development of NEC lesions and confirm the enteropathogenicity of Clostridium butyricum and Clostridium neonatale in NEC. First, we produced C.butyricum and C.neonatale strains impaired in butyrate production by genetically inactivating the hbd gene encoding ß-hydroxybutyryl-CoA dehydrogenase that produces end-fermentation metabolites. Second, we evaluated the enteropathogenicty of the hbd-knockout strains in a gnotobiotic quail model of NEC. The analyses showed that animals harboring these strains had significantly fewer and less intense intestinal lesions than those harboring the respective wild-type strains. In the absence of specific biological markers of NEC, the data provide original and new mechanistic insights into the disease pathophysiology, a necessary step for developing potential novel therapies.


Subject(s)
Clostridium butyricum , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Newborn, Diseases , Infant, Newborn , Humans , Animals , Clostridium butyricum/genetics , Enterocolitis, Necrotizing/microbiology , Fermentation , Butyrates
19.
Nutrients ; 15(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36771414

ABSTRACT

The development and health of infants are intertwined with the protective and regulatory functions of different microorganisms in the gut known as the gut microbiota. Preterm infants born with an imbalanced gut microbiota are at substantial risk of several diseases including inflammatory intestinal diseases, necrotizing enterocolitis, late-onset sepsis, neurodevelopmental disorders, and allergies which can potentially persist throughout adulthood. In this review, we have evaluated the role of Bifidobacterium as commonly used probiotics in the development of gut microbiota and prevention of common diseases in preterm infants which is not fully understood yet. The application of Bifidobacterium as a therapeutical approach in the re-programming of the gut microbiota in preterm infants, the mechanisms of host-microbiome interaction, and the mechanism of action of this bacterium have also been investigated, aiming to provide new insights and opportunities in microbiome-targeted interventions in personalized medicine.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Probiotics , Sepsis , Infant , Infant, Newborn , Humans , Adult , Infant, Premature , Bifidobacterium , Sepsis/prevention & control , Probiotics/therapeutic use , Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/microbiology
20.
Nutrients ; 14(22)2022 Nov 12.
Article in English | MEDLINE | ID: mdl-36432481

ABSTRACT

Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease in premature infants and a leading cause of death in neonates (1-7% in the US). NEC is caused by opportunistic bacteria, which cause gut dysbiosis and inflammation and ultimately result in intestinal necrosis. Previous studies have utilized the rodent and pig models to mimic NEC, whereas the current study uses the in vivo (Gallus gallus) intra-amniotic administration approach to investigate NEC. On incubation day 17, broiler chicken (Gallus gallus) viable embryos were injected intra-amniotically with 1 mL dextran sodium sulfate (DSS) in H2O. Four treatment groups (0.1%, 0.25%, 0.5%, and 0.75% DSS) and two controls (H2O/non-injected controls) were administered. We observed a significant increase in intestinal permeability and negative intestinal morphological changes, specifically, decreased villus surface area and goblet cell diameter in the 0.50% and 0.75% DSS groups. Furthermore, there was a significant increase in pathogenic bacterial (E. coli spp. and Klebsiella spp.) abundances in the 0.75% DSS group compared to the control groups, demonstrating cecal microbiota dysbiosis. These results demonstrate significant physiopathology of NEC and negative bacterial-host interactions within a premature gastrointestinal system. Our present study demonstrates a novel model of NEC through intra-amniotic administration to study the effects of NEC on intestinal functionality, morphology, and gut microbiota in vivo.


Subject(s)
Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Infant, Newborn , Humans , Female , Animals , Swine , Enterocolitis, Necrotizing/microbiology , Chickens , Dysbiosis , Escherichia coli , Bacteria
SELECTION OF CITATIONS
SEARCH DETAIL