Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Newborn (Clarksville) ; 1(2): 219-226, 2022.
Article in English | MEDLINE | ID: mdl-36237439

ABSTRACT

Early life microbial colonization is critical for the development of the immune system, postnatal growth, and long-term health and disease. The dynamic and nascent microbiomes of children are highly individualized and are characterized by low bacterial diversity. Any disruptions in microbial colonization can contribute to shifts in normal microbial colonization that persist past the first 1000 days of life and result in intestinal dysbiosis. Here, we focus on microbiome-host interactions during fetal, newborn, and infant microbiome development. We summarize the roles of bacterial communities in fetal development and adverse health outcomes due to dysbiosis. We also discuss how internal and external factors program the microbiome's metabolic machinery as it evolves into an adult-like microbiome. Finally, we discuss the limits of current studies and future directions. Studies on the early-life microbiome will be critical for a better understanding of childhood health and diseases, as well as restorative methods for the prevention and treatment of diseases in adulthood.

2.
Semin Perinatol ; 46(1): 151539, 2022 02.
Article in English | MEDLINE | ID: mdl-34887106

ABSTRACT

In the setting of threatened extreme preterm birth, balancing maternal and fetal risks and benefits in order to choose the best available treatment options is of utmost importance. Inconsistency in treatment practices for infants born between 22 and 24 weeks of gestatotional age may account for inter-hospital variation in survival rates with and without impairment. Most importantly, non-biased and accurate information must be presented to the family as soon as extremely preterm birth is suspected, including counseling on morbidities and mortality associated with delivery at the limits of viability. This review will focus on different therapeutic medical and surgical practices available for threatened extremely preterm birth to improve fetal and maternal outcomes while highlighting the importance of patient-centered approaches.


Subject(s)
Mothers , Premature Birth , Female , Gestational Age , Humans , Infant , Infant, Newborn , Morbidity , Pregnancy , Prenatal Care
3.
Gut Microbes ; 13(1): 1-7, 2021.
Article in English | MEDLINE | ID: mdl-33960272

ABSTRACT

Mortality and morbidity from SARS-CoV2 (COVID-19) infections in children remains low, including an exceedingly low rate of horizontal and vertical transmission. However, unforeseen complications to childhood health have emerged secondary to the pandemic. Few studies to date have examined unintended complications of the pandemic in newborns and infants. In this Commentary, we discuss the impact that COVID-19 may have on inheritance of the newborn microbiome and its assembly throughout the first years of life. In the early stages of the pandemic when vertical transmission of COVID-19 was poorly understood, several studies reported increased rates of C-sections in COVID-19 positive women. Initial recommendations discouraged COVID-19 positive mothers from breastfeeding and participating in skin-to-skin care, advising them to isolate during their window of infectivity. These shifts in perinatal care can adversely impact microbial colonization during the first 1000 days of life. While obstetrical and neonatal management have evolved to reflect our current knowledge of perinatal transmission, we are observing other changes in early life exposures of infants, including increased attention to hygiene, fewer social interactions, and decreased global travel, all of which are major drivers of early-life gut colonization. Composition of the gut microbiota in adults directly impacts severity of infection, suggesting a role of microbial communities in modulating immune responses to COVID-19. Conversely, the role of the intestinal microbiome in susceptibility and severity of COVID-19 in newborns and children remains unknown. The onset of adulthood diseases is related to the establishment of a healthy gut microbiome during childhood. As we continue to define COVID-19 biology, further research is necessary to understand how acquisition of the neonatal microbiome is affected by the pandemic. Furthermore, infection control measures must be balanced with strategies that promote microbial diversity to impart optimal health outcomes and potentially modulate susceptibility of children to COVID-19.


Subject(s)
COVID-19/complications , COVID-19/physiopathology , Gastrointestinal Microbiome/physiology , Infectious Disease Transmission, Vertical , Microbiota/physiology , Pregnancy Complications, Infectious/etiology , Pregnancy Complications, Infectious/physiopathology , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Pregnancy , SARS-CoV-2
4.
Gastroenterol Clin North Am ; 50(1): 77-100, 2021 03.
Article in English | MEDLINE | ID: mdl-33518170

ABSTRACT

Bacterial colonization of the intestines occurs during the first 2 years of life. Homeostasis of the gut microbiome is established to foster normal intestinal immune development for adulthood. Derangements in this process can interfere with immune function and increase an individual's risk for gastrointestinal disorders. We discuss the role of diet and the microbiome on the onset of such disorders. We examine how micronutrients, prebiotics, and probiotics modulate disease pathogenesis. We discuss how diet and abnormal microbial colonization impact extraintestinal organs. Understanding the communication of nutrition and the microbiome offers exciting opportunities for therapeutics.


Subject(s)
Gastrointestinal Diseases , Gastrointestinal Microbiome , Probiotics , Adult , Humans , Intestines , Prebiotics , Probiotics/therapeutic use
5.
medRxiv ; 2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33442705

ABSTRACT

Perinatal transmission of COVID-19 is poorly understood and many neonatal intensive care units' (NICU) policies minimize mother-infant contact to prevent transmission. We present our unit's approach and ways it may impact neonatal microbiome acquisition. We attended COVID-19 positive mothers' deliveries from March-August 2020. Delayed cord clamping and skin-to-skin were avoided and infants were admitted to the NICU. No parents' visits were allowed and discharge was arranged with COVID-19 negative family members. Maternal breast milk was restricted in the NICU. All twenty-one infants tested negative at 24 and 48 hours and had average hospital stays of nine days. 40% of mothers expressed breastmilk and 60% of infants were discharged with COVID-19 negative caregivers. Extended hospital stays, no skin-to-skin contact, limited maternal milk use, and discharge to caregivers outside primary residences, potentially affect the neonatal microbiome. Future studies are warranted to explore how ours and other centers' similar policies influence this outcome.

6.
PLoS One ; 13(10): e0206366, 2018.
Article in English | MEDLINE | ID: mdl-30365522

ABSTRACT

OBJECTIVE: Necrotizing enterocolitis (NEC) is the most common surgical emergency in preterm infants, and pathogenesis associates with changes in the fecal microbiome. As fecal samples incompletely represent microbial communities in intestinal mucosa, we sought to determine the NEC tissue-specific microbiome and assess its contribution to pathogenesis. DESIGN: We amplified and sequenced the V1-V3 hypervariable region of the bacterial 16S rRNA gene extracted from intestinal tissue and corresponding fecal samples from 12 surgical patients with NEC and 14 surgical patients without NEC. Low quality and non-bacterial sequences were removed, and taxonomic assignment was made with the Ribosomal Database Project. Operational taxonomic units were clustered at 97%. We tested for differences between NEC and non-NEC samples in microbiome alpha- and beta-diversity and differential abundance of specific taxa between NEC and non-NEC samples. Additional analyses were performed to assess the contribution of other demographic and environmental confounding factors on the infant tissue and fecal microbiome. RESULTS: The fecal and tissue microbial communities were different. NEC was associated with a distinct microbiome, which was characterized by low diversity, higher abundances of Staphylococcus and Clostridium_sensu_stricto, and lower abundances of Actinomyces and Corynebacterium. Infant age and vancomycin exposure correlated with shifts in the tissue microbiome. CONCLUSION: The observed low diversity in NEC tissues suggests that NEC is associated with a bacterial bloom and a distinct mucosal bacterial community. The exact bacterial species that constitute the bloom varied by infant and were strongly influenced by age and exposure to vancomycin.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/surgery , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Age Factors , Anti-Bacterial Agents/pharmacology , Biodiversity , Enterocolitis, Necrotizing/drug therapy , Female , Humans , Infant , Infant, Newborn , Male , Microbiota/drug effects , Pregnancy
7.
PLoS One ; 13(5): e0197439, 2018.
Article in English | MEDLINE | ID: mdl-29771989

ABSTRACT

Bacterial DNA has been reported in the placenta and amniotic fluid by several independent groups of investigators. However, it's taxonomic overlap with fetal and maternal bacterial DNA in different sites has been poorly characterized. Here, we determined the presence of bacterial DNA in the intestines and placentas of fetal mice at gestational day 17 (n = 13). These were compared to newborn intestines (n = 15), maternal sites (mouth, n = 6; vagina, n = 6; colon, n = 7; feces, n = 8), and negative controls to rule out contamination. The V4 region of the bacterial 16S rRNA gene indicated a pattern of bacterial DNA in fetal intestine similar to placenta but with higher phylogenetic diversity than placenta or newborn intestine. Firmicutes were the most frequently assignable phylum. SourceTracker analysis suggested the placenta as the most commonly identifiable origin for fetal bacterial DNA, but also over 75% of fetal gut genera overlapped with maternal oral and vaginal taxa but not with maternal or newborn feces. These data provide evidence for the presence of bacterial DNA in the mouse fetus.


Subject(s)
Amniotic Fluid/metabolism , DNA, Bacterial/analysis , Intestinal Mucosa/metabolism , Intestines/embryology , Placenta/metabolism , Placenta/microbiology , Animals , Female , Mice , Pregnancy , RNA, Ribosomal, 16S/genetics , Vagina/metabolism , Vagina/microbiology
8.
Article in English | MEDLINE | ID: mdl-28217556

ABSTRACT

Streptococcus agalactiae, or Group B Streptococcus (GBS), is a gram-positive bacterial pathogen associated with infection during pregnancy and is a major cause of morbidity and mortality in neonates. Infection of the extraplacental membranes surrounding the developing fetus, a condition known as chorioamnionitis, is characterized histopathologically by profound infiltration of polymorphonuclear cells (PMNs, neutrophils) and greatly increases the risk for preterm labor, stillbirth, or neonatal GBS infection. The advent of animal models of chorioamnionitis provides a powerful tool to study host-pathogen relationships in vivo and ex vivo. The purpose of this study was to evaluate the innate immune response elicited by GBS and evaluate how antimicrobial strategies elaborated by these innate immune cells affect bacteria. Our work using a mouse model of GBS ascending vaginal infection during pregnancy reveals that clinically isolated GBS has the capacity to invade reproductive tissues and elicit host immune responses including infiltration of PMNs within the choriodecidua and placenta during infection, mirroring the human condition. Upon interacting with GBS, murine neutrophils elaborate DNA-containing extracellular traps, which immobilize GBS and are studded with antimicrobial molecules including lactoferrin. Exposure of GBS to holo- or apo-forms of lactoferrin reveals that the iron-sequestration activity of lactoferrin represses GBS growth and viability in a dose-dependent manner. Together, these data indicate that the mouse model of ascending infection is a useful tool to recapitulate human models of GBS infection during pregnancy. Furthermore, this work reveals that neutrophil extracellular traps ensnare GBS and repress bacterial growth via deposition of antimicrobial molecules, which drive nutritional immunity via metal sequestration strategies.


Subject(s)
Extracellular Traps , Immunity, Innate , Mucous Membrane/pathology , Neutrophil Infiltration , Reproductive Tract Infections/pathology , Streptococcus agalactiae/pathogenicity , Animals , Anti-Bacterial Agents/metabolism , Disease Models, Animal , Female , Iron/metabolism , Lactoferrin/metabolism , Mice , Microbial Viability/drug effects , Mucous Membrane/immunology , Reproductive Tract Infections/immunology , Streptococcus agalactiae/immunology
9.
J Immunol ; 195(3): 1064-70, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26101326

ABSTRACT

Sepsis is a major cause of neonatal mortality and morbidity worldwide. A recent report suggested that murine neonatal host defense against infection could be compromised by immunosuppressive CD71(+) erythroid splenocytes. We examined the impact of CD71(+) erythroid splenocytes on murine neonatal mortality to endotoxin challenge or polymicrobial sepsis and characterized circulating CD71(+) erythroid (CD235a(+)) cells in human neonates. Adoptive transfer or an Ab-mediated reduction in neonatal CD71(+) erythroid splenocytes did not alter murine neonatal survival to endotoxin challenge or polymicrobial sepsis challenge. Ex vivo immunosuppression of stimulated adult CD11b(+) cells was not limited to neonatal splenocytes; it also occurred with adult and neonatal bone marrow. Animals treated with anti-CD71 Ab showed reduced splenic bacterial load following bacterial challenge compared with isotype-treated mice. However, adoptive transfer of enriched CD71(+) erythroid splenocytes to CD71(+)-reduced animals did not reduce bacterial clearance. Human CD71(+)CD235a(+) cells were common among cord blood mononuclear cells and were shown to be reticulocytes. In summary, a lack of effect on murine survival to polymicrobial sepsis following adoptive transfer or diminution of CD71(+) erythroid splenocytes under these experimental conditions suggests that the impact of these cells on neonatal infection risk and progression may be limited. An unanticipated immune priming effect of anti-CD71 Ab treatment, rather than a reduction in immunosuppressive CD71(+) erythroid splenocytes, was likely responsible for the reported enhanced bacterial clearance. In humans, the well-described rapid decrease in circulating reticulocytes after birth suggests that they may have a limited role in reducing inflammation secondary to microbial colonization.


Subject(s)
Antigens, CD/immunology , Bone Marrow Cells/immunology , Erythroid Cells/immunology , Receptors, Transferrin/immunology , Sepsis/immunology , Adoptive Transfer , Animals , Antibodies/immunology , CD11b Antigen/metabolism , Endotoxins/pharmacology , Female , Fetal Blood/cytology , Fetal Blood/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Reticulocytes/immunology , Spleen/cytology , Spleen/immunology
10.
Pediatr Res ; 77(1-2): 189-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25310759

ABSTRACT

While critical for normal development, the exact timing of establishment of the intestinal microbiome is unknown. For example, although preterm labor and birth have been associated with bacterial colonization of the amniotic cavity and fetal membranes for many years, the prevailing dogma of a sterile intrauterine environment during normal term pregnancies has been challenged more recently. While found to be a key contributor of evolution in the animal kingdom, maternal transmission of commensal bacteria may also constitute a critical process during healthy pregnancies in humans with yet unclear developmental importance. Metagenomic sequencing has elucidated a rich placental microbiome in normal term pregnancies likely providing important metabolic and immune contributions to the growing fetus. Conversely, an altered microbial composition during pregnancy may produce aberrant metabolites impairing fetal brain development and life-long neurological outcomes. Here we review the current understanding of microbial colonization at the feto-maternal interface and explain how normal gut colonization drives a balanced neonatal mucosal immune system, while dysbiosis contributes to aberrant immune function early in life and beyond. We discuss how maternal genetics, diet, medications, and probiotics inform the fetal microbiome in preparation for perinatal and postnatal bacterial colonization.


Subject(s)
Fetus/immunology , Fetus/microbiology , Immune System/embryology , Maternal-Fetal Exchange/immunology , Microbiota/immunology , Models, Immunological , Placenta/microbiology , Female , Humans , Immune System/growth & development , Pregnancy
11.
Chest ; 147(6): 1494-1502, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25474571

ABSTRACT

BACKGROUND: Ventilator-associated pneumonia (VAP) remains a common complication in critically ill surgical patients, and its diagnosis remains problematic. Exhaled breath contains aerosolized droplets that reflect the lung microbiota. We hypothesized that exhaled breath condensate fluid (EBCF) in hygroscopic condenser humidifier/heat and moisture exchanger (HCH/HME) filters would contain bacterial DNA that qualitatively and quantitatively correlate with pathogens isolated from quantitative BAL samples obtained for clinical suspicion of pneumonia. METHODS: Forty-eight adult patients who were mechanically ventilated and undergoing quantitative BAL (n = 51) for suspected pneumonia in the surgical ICU were enrolled. Per protocol, patients fulfilling VAP clinical criteria undergo quantitative BAL bacterial culture. Immediately prior to BAL, time-matched HCH/HME filters were collected for study of EBCF by real-time polymerase chain reaction. Additionally, convenience samples of serially collected filters in patients with BAL-diagnosed VAP were analyzed. RESULTS: Forty-nine of 51 time-matched EBCF/BAL fluid samples were fully concordant (concordance > 95% by κ statistic) relative to identified pathogens and strongly correlated with clinical cultures. Regression analysis of quantitative bacterial DNA in paired samples revealed a statistically significant positive correlation (r = 0.85). In a convenience sample, qualitative and quantitative polymerase chain reaction analysis of serial HCH/HME samples for bacterial DNA demonstrated an increase in load that preceded the suspicion of pneumonia. CONCLUSIONS: Bacterial DNA within EBCF demonstrates a high correlation with BAL fluid and clinical cultures. Bacterial DNA within EBCF increases prior to the suspicion of pneumonia. Further study of this novel approach may allow development of a noninvasive tool for the early diagnosis of VAP.


Subject(s)
Diagnostic Tests, Routine/methods , Exhalation , Lung/microbiology , Microbiological Techniques/methods , Microbiota/genetics , Pneumonia, Ventilator-Associated/diagnosis , Pneumonia, Ventilator-Associated/microbiology , Bronchoalveolar Lavage/instrumentation , Bronchoalveolar Lavage/methods , Critical Illness , DNA, Bacterial/genetics , Diagnostic Tests, Routine/instrumentation , Humans , Intensive Care Units , Microbiological Techniques/instrumentation , Pilot Projects , Real-Time Polymerase Chain Reaction , Regression Analysis , Sensitivity and Specificity
12.
Gut Microbes ; 5(2): 192-201, 2014.
Article in English | MEDLINE | ID: mdl-24637795

ABSTRACT

Fecal sampling is widely utilized to define small intestinal tissue-level microbial communities in healthy and diseased newborns. However, this approach may lead to inaccurate assessments of disease or therapeutics in newborns because of the assumption that the taxa in the fecal microbiota are representative of the taxa present throughout the gastrointestinal tract. To assess the stratification of microbes in the newborn gut and to evaluate the probable shortcoming of fecal sampling in place of tissue sampling, we simultaneously compared intestinal mucosa and fecal microbial communities in 15 neonates undergoing intestinal resections. We report three key results. First, when the site of fecal and mucosal samples are further apart, their microbial communities are more distinct, as indicated by low mean Sørensen similarity indices for each patient's fecal and tissue microbiota. Second, two distinct niches (intestinal mucosa and fecal microbiota) are evident by principal component analyses, demonstrating the critical role of sample source in defining microbial composition. Finally, in contrast to adult studies, intestinal bacterial diversity was higher in tissue than in fecal samples. This study represents an unprecedented map of the infant microbiota from intestinal mucosa and establishes discernable biogeography throughout the neonatal gastrointestinal tract. Our results question the reliance on fecal microbiota as a proxy for the developing intestinal microbiota. Additionally, the robust intestinal tissue-level bacterial diversity we detected at these early ages may contribute to the maturation of mucosal immunity.


Subject(s)
Gastrointestinal Tract/microbiology , Bacteria/genetics , Bacteria/isolation & purification , Feces/microbiology , Humans , Infant, Newborn , Intestinal Mucosa/microbiology
13.
J Am Med Inform Assoc ; 21(2): 326-36, 2014.
Article in English | MEDLINE | ID: mdl-24043317

ABSTRACT

OBJECTIVE: The objective was to develop non-invasive predictive models for late-onset neonatal sepsis from off-the-shelf medical data and electronic medical records (EMR). DESIGN: The data used in this study are from 299 infants admitted to the neonatal intensive care unit in the Monroe Carell Jr. Children's Hospital at Vanderbilt and evaluated for late-onset sepsis. Gold standard diagnostic labels (sepsis negative, culture positive sepsis, culture negative/clinical sepsis) were assigned based on all the laboratory, clinical and microbiology data available in EMR. Only data that were available up to 12 h after phlebotomy for blood culture testing were used to build predictive models using machine learning (ML) algorithms. MEASUREMENT: We compared sensitivity, specificity, positive predictive value and negative predictive value of sepsis treatment of physicians with the predictions of models generated by ML algorithms. RESULTS: The treatment sensitivity of all the nine ML algorithms and specificity of eight out of the nine ML algorithms tested exceeded that of the physician when culture-negative sepsis was included. When culture-negative sepsis was excluded both sensitivity and specificity exceeded that of the physician for all the ML algorithms. The top three predictive variables were the hematocrit or packed cell volume, chorioamnionitis and respiratory rate. CONCLUSIONS: Predictive models developed from off-the-shelf and EMR data using ML algorithms exceeded the treatment sensitivity and treatment specificity of clinicians. A prospective study is warranted to assess the clinical utility of the ML algorithms in improving the accuracy of antibiotic use in the management of neonatal sepsis.


Subject(s)
Algorithms , Artificial Intelligence , Diagnosis, Computer-Assisted , Electronic Health Records , Sepsis/diagnosis , Anti-Bacterial Agents/therapeutic use , Decision Support Techniques , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Sensitivity and Specificity , Sepsis/drug therapy
14.
Curr Opin Endocrinol Diabetes Obes ; 19(2): 73-80, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22357099

ABSTRACT

PURPOSE OF REVIEW: The microbiome continues to demonstrate an important role in immune and metabolic programming. This review will focus on the mechanistic implications of recent findings for diabetes pathogenesis and treatment. RECENT FINDINGS: Multiple techniques are developing to specify the microbiome. At the same time, new insights have emerged into local interactions of microbial products with human development. New findings demonstrate that key bacteria and their products result in the programming of diabetes-modulating Th17 and regulatory T lymphocytes within and outside the intestine. The role of the bacterial metagenome in programming human metabolism has also revealed new insights. In turn, these findings suggest a framework in which the microbiome may be modified to change the course of diabetes. SUMMARY: The microbiome is a key regulator of metabolism and immunity. Specific bacteria and their secreted products are now known to program Th17 and regulatory T-cell development, which may change the course of diabetes. Bacterial genomics are demonstrating important, modifiable roles of bacterial gene products in metabolism. Further understanding of this symbiotic relationship will provide new avenues for intervention in diabetes.


Subject(s)
Adaptive Immunity/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/immunology , Intestines/microbiology , Metagenome/immunology , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 2/microbiology , Female , Humans , Male , T-Lymphocytes, Regulatory
SELECTION OF CITATIONS
SEARCH DETAIL
...