131 results on '"Michael J. Morowitz"'
Search Results
2. Using strain-resolved analysis to identify contamination in metagenomics data
- Author
-
Yue Clare Lou, Jordan Hoff, Matthew R. Olm, Jacob West-Roberts, Spencer Diamond, Brian A. Firek, Michael J. Morowitz, and Jillian F. Banfield
- Subjects
Contamination ,Microbiome ,Strains ,Genome-resolved metagenomics ,Microbial ecology ,QR100-130 - Abstract
Abstract Background Metagenomics analyses can be negatively impacted by DNA contamination. While external sources of contamination such as DNA extraction kits have been widely reported and investigated, contamination originating within the study itself remains underreported. Results Here, we applied high-resolution strain-resolved analyses to identify contamination in two large-scale clinical metagenomics datasets. By mapping strain sharing to DNA extraction plates, we identified well-to-well contamination in both negative controls and biological samples in one dataset. Such contamination is more likely to occur among samples that are on the same or adjacent columns or rows of the extraction plate than samples that are far apart. Our strain-resolved workflow also reveals the presence of externally derived contamination, primarily in the other dataset. Overall, in both datasets, contamination is more significant in samples with lower biomass. Conclusion Our work demonstrates that genome-resolved strain tracking, with its essentially genome-wide nucleotide-level resolution, can be used to detect contamination in sequencing-based microbiome studies. Our results underscore the value of strain-specific methods to detect contamination and the critical importance of looking for contamination beyond negative and positive controls. Video Abstract
- Published
- 2023
- Full Text
- View/download PDF
3. Epithelial NAD+ depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
- Author
-
Elizabeth A. Novak, Erin C. Crawford, Heather L. Mentrup, Brian D. Griffith, David M. Fletcher, Meredith R. Flanagan, Corinne Schneider, Brian Firek, Matthew B. Rogers, Michael J. Morowitz, Jon D. Piganelli, Qian Wang, and Kevin P. Mollen
- Subjects
colitis ,PGC1α ,nicotinamide riboside ,poly(ADP) riboside polymers ,nicotinamide adenine dinucleotide ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionWe have previously demonstrated that a pathologic downregulation of peroxisome proliferator-activated receptor–gamma coactivator 1-alpha (PGC1α) within the intestinal epithelium contributes to the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism underlying downregulation of PGC1α expression and activity during IBD is not yet clear.MethodsMice (male; C57Bl/6, Villincre/+;Pgc1afl/fl mice, and Pgc1afl/fl) were subjected to experimental colitis and treated with nicotinamide riboside. Western blot, high-resolution respirometry, nicotinamide adenine dinucleotide (NAD+) quantification, and immunoprecipitation were used to in this study.ResultsWe demonstrate a significant depletion in the NAD+ levels within the intestinal epithelium of mice undergoing experimental colitis, as well as humans with ulcerative colitis. While we found no decrease in the levels of NAD+-synthesizing enzymes within the intestinal epithelium of mice undergoing experimental colitis, we did find an increase in the mRNA level, as well as the enzymatic activity, of the NAD+-consuming enzyme poly(ADP-ribose) polymerase-1 (PARP1). Treatment of mice undergoing experimental colitis with an NAD+ precursor reduced the severity of colitis, restored mitochondrial function, and increased active PGC1α levels; however, NAD+ repletion did not benefit transgenic mice that lack PGC1α within the intestinal epithelium, suggesting that the therapeutic effects require an intact PGC1α axis.DiscussionOur results emphasize the importance of PGC1α expression to both mitochondrial health and homeostasis within the intestinal epithelium and suggest a novel therapeutic approach for disease management. These findings also provide a mechanistic basis for clinical trials of nicotinamide riboside in IBD patients.
- Published
- 2023
- Full Text
- View/download PDF
4. Crosstalk between DNA Damage Repair and Metabolic Regulation in Hematopoietic Stem Cells
- Author
-
Jian Xu, Peiwen Fei, Dennis W. Simon, Michael J. Morowitz, Parinda A. Mehta, and Wei Du
- Subjects
hematopoietic stem cells (HSCs) ,DNA damage repair (DDR) ,cellular metabolism ,Fanconi anemia (FA) pathway ,Cytology ,QH573-671 - Abstract
Self-renewal and differentiation are two characteristics of hematopoietic stem cells (HSCs). Under steady physiological conditions, most primitive HSCs remain quiescent in the bone marrow (BM). They respond to different stimuli to refresh the blood system. The transition from quiescence to activation is accompanied by major changes in metabolism, a fundamental cellular process in living organisms that produces or consumes energy. Cellular metabolism is now considered to be a key regulator of HSC maintenance. Interestingly, HSCs possess a distinct metabolic profile with a preference for glycolysis rather than oxidative phosphorylation (OXPHOS) for energy production. Byproducts from the cellular metabolism can also damage DNA. To counteract such insults, mammalian cells have evolved a complex and efficient DNA damage repair (DDR) system to eliminate various DNA lesions and guard genomic stability. Given the enormous regenerative potential coupled with the lifetime persistence of HSCs, tight control of HSC genome stability is essential. The intersection of DDR and the HSC metabolism has recently emerged as an area of intense research interest, unraveling the profound connections between genomic stability and cellular energetics. In this brief review, we delve into the interplay between DDR deficiency and the metabolic reprogramming of HSCs, shedding light on the dynamic relationship that governs the fate and functionality of these remarkable stem cells. Understanding the crosstalk between DDR and the cellular metabolism will open a new avenue of research designed to target these interacting pathways for improving HSC function and treating hematologic disorders.
- Published
- 2024
- Full Text
- View/download PDF
5. Experimental validation that human microbiome phages use alternative genetic coding
- Author
-
Samantha L. Peters, Adair L. Borges, Richard J. Giannone, Michael J. Morowitz, Jillian F. Banfield, and Robert L. Hettich
- Subjects
Science - Abstract
Previous bioinformatic analyses have indicated that bacteriophages can use genetic codes different from those of their host bacteria. Here, Peters et al. use metaproteomics to provide experimental evidence of reassignment of stop codon TAG to glutamine in phages found in the human gut microbiome.
- Published
- 2022
- Full Text
- View/download PDF
6. The NICU Antibiotics and Outcomes (NANO) trial: a randomized multicenter clinical trial assessing empiric antibiotics and clinical outcomes in newborn preterm infants
- Author
-
Michael J. Morowitz, Anup C. Katheria, Richard A. Polin, Elizabeth Pace, David T. Huang, Chung-Chou H. Chang, and Johathan G. Yabes
- Subjects
Microbial colonization ,Extremely-low-birthweight ,Prematurity ,Early-onset neonatal sepsis ,Late-onset neonatal sepsis ,Necrotizing enterocolitis ,Medicine (General) ,R5-920 - Abstract
Abstract Background Early-onset sepsis is an important cause of neonatal morbidity and mortality in the preterm population. Infants perceived to be at increased risk for early-onset sepsis are often treated empirically with broad-spectrum antibiotics while awaiting confirmatory blood cultures, despite an overall incidence of early-onset sepsis of 2–3% among extremely-low-birthweight (ELBW) infants. Recent observational studies associate perinatal antibiotic use with an increased incidence of necrotizing enterocolitis, late-onset sepsis, and mortality among ELBW infants. Given currently available data and variability in clinical practice, we designed a prospective multi-institutional randomized controlled trial to determine the safety of early antibiotic use in ELBW infants. Methods The NICU Antibiotics and Outcomes (NANO) trial is a multicenter, double-blinded, randomized controlled trial. A sample of 802 ELBW preterm infants will undergo web-based stratified block randomization to receive empiric antibiotics (EA; ampicillin and gentamicin) or placebo during routine evaluation for early-onset sepsis. Participating sites will use preexisting institutional protocols for antibiotic dosage and duration. Infants born at participating sites with a gestational age of 29 weeks or less are eligible for enrollment. Exclusion criteria include maternal intrauterine infection, hemodynamic or respiratory instability, delivery by caesarean section for maternal indications without labor or prolonged rupture of membranes, and prior administration of antibiotics. The primary outcome is the composite incidence of necrotizing enterocolitis, late-onset sepsis, or death during participants’ index hospitalization. Maternal and infant samples will be collected longitudinally and assessed for differences in microbiome composition and diversity. Discussion The NANO trial is designed to compare the rate of adverse outcomes of EA use at birth versus placebo in ELBW preterm infants. If EA at birth worsens clinical outcomes, then the results of the trial may help providers decrease antibiotic utilization in the NICU and subsequently decrease the incidence of complications associated with early antibiotic use in ELBW infants. If we instead find that EA improve outcomes, then the trial will validate a longstanding clinical practice that has not previously been supported by high-quality data. Future studies will assess long-term clinical and microbial outcomes in infants who received empiric antibiotics following delivery. Trial registration Trial registration data: June 25, 2019 NCT03997266 .
- Published
- 2022
- Full Text
- View/download PDF
7. Early antibiotics and risk for necrotizing enterocolitis in premature infants: A narrative review
- Author
-
Alain Cuna, Michael J. Morowitz, and Venkatesh Sampath
- Subjects
antibiotic stewardship ,intestinal microbiome ,prematurity ,necrotizing entercolitis ,antibiotics ,postnatal intestinal adaptation ,Pediatrics ,RJ1-570 - Abstract
While prompt initiation of antibiotics at birth due to concerns for early onset sepsis is common, it often leads to many preterm infants being exposed to treatment despite negative blood cultures. Such exposure to early antibiotics can impact the developing gut microbiome putting infants at increased risk of several diseases. Necrotizing enterocolitis (NEC), a devastating inflammatory bowel disease that affects preterm infants, is among the most widely studied neonatal disease that has been linked to early antibiotics. While some studies have demonstrated an increased risk of NEC, other studies have demonstrated seemingly contrary findings of decreased NEC with early antibiotics. Studies using animal models have also yielded differing findings of benefit vs. harm of early antibiotic exposure on subsequent NEC susceptibility. We thus sought to conduct this narrative review to help clarify the relationship between early antibiotics exposure and future risk of NEC in preterm infants. Our objectives are to: (1) summarize findings from human and animal studies that investigated the relationship between early antibiotics and NEC, (2) highlight important limitations of these studies, (3) explore potential mechanisms that can explain why early antibiotics may increase or decrease NEC risk, and (4) identify future directions for research.
- Published
- 2023
- Full Text
- View/download PDF
8. Antibiotic resistance and host immune system-induced metal bactericidal control are key factors for microbial persistence in the developing human preterm infant gut microbiome
- Author
-
Samantha L. Peters, Michael J. Morowitz, and Robert L. Hettich
- Subjects
human microbiome ,human infant faecal microbiota ,Metaproteomics ,host immune system ,metal bactericidal control ,antibiotic resistance ,Microbiology ,QR1-502 - Abstract
The human gut microbiome, which develops and stabilizes during the early stages of infant life, plays an essential role in host health through the production of metabolic resources and the stimulation and training of the immune system. To study colonization and community functional dynamics of the microbiota based on responses to host immune processes during the normal and dysbiotic establishment of the gut, metaproteomics was conducted on 91 fecal samples collected over the first 90 days of life from 17 hospitalized premature infants. Microbial responses to antibiotic administration and host-imposed metal bactericidal control correlated with community assembly and resiliency of microbes in the developing preterm gut. Specifically, proteins related to antibiotic resistance and metal homeostasis mechanisms were predominant in persisting members in the infant gut environment over the first several weeks of life. Overall, this metaproteomics study provides a unique approach to examine the temporal expansion and resilience of microbial colonization, as it allows simultaneous examination of both host and microbial metabolic activities. Understanding the interplay between host and microbes may elucidate the microbiome’s potential immunomodulatory roles relevant to necrotizing enterocolitis and other dysbiotic conditions in preterm infants.
- Published
- 2022
- Full Text
- View/download PDF
9. Genetic and behavioral adaptation of Candida parapsilosis to the microbiome of hospitalized infants revealed by in situ genomics, transcriptomics, and proteomics
- Author
-
Patrick T. West, Samantha L. Peters, Matthew R. Olm, Feiqiao B. Yu, Haley Gause, Yue Clare Lou, Brian A. Firek, Robyn Baker, Alexander D. Johnson, Michael J. Morowitz, Robert L. Hettich, and Jillian F. Banfield
- Subjects
Microbial eukaryotes ,Metagenomics ,Genome-resolved metagenomics ,Strain-tracking ,Hospital microbiome ,Neonatal intensive care unit ,Microbial ecology ,QR100-130 - Abstract
Abstract Background Candida parapsilosis is a common cause of invasive candidiasis, especially in newborn infants, and infections have been increasing over the past two decades. C. parapsilosis has been primarily studied in pure culture, leaving gaps in understanding of its function in a microbiome context. Results Here, we compare five unique C. parapsilosis genomes assembled from premature infant fecal samples, three of which are newly reconstructed, and analyze their genome structure, population diversity, and in situ activity relative to reference strains in pure culture. All five genomes contain hotspots of single nucleotide variants, some of which are shared by strains from multiple hospitals. A subset of environmental and hospital-derived genomes share variants within these hotspots suggesting derivation of that region from a common ancestor. Four of the newly reconstructed C. parapsilosis genomes have 4 to 16 copies of the gene RTA3, which encodes a lipid translocase and is implicated in antifungal resistance, potentially indicating adaptation to hospital antifungal use. Time course metatranscriptomics and metaproteomics on fecal samples from a premature infant with a C. parapsilosis blood infection revealed highly variable in situ expression patterns that are distinct from those of similar strains in pure cultures. For example, biofilm formation genes were relatively less expressed in situ, whereas genes linked to oxygen utilization were more highly expressed, indicative of growth in a relatively aerobic environment. In gut microbiome samples, C. parapsilosis co-existed with Enterococcus faecalis that shifted in relative abundance over time, accompanied by changes in bacterial and fungal gene expression and proteome composition. Conclusions The results reveal potentially medically relevant differences in Candida function in gut vs. laboratory environments, and constrain evolutionary processes that could contribute to hospital strain persistence and transfer into premature infant microbiomes. Video abstract
- Published
- 2021
- Full Text
- View/download PDF
10. Infant gut strain persistence is associated with maternal origin, phylogeny, and traits including surface adhesion and iron acquisition
- Author
-
Yue Clare Lou, Matthew R. Olm, Spencer Diamond, Alexander Crits-Christoph, Brian A. Firek, Robyn Baker, Michael J. Morowitz, and Jillian F. Banfield
- Subjects
strain-resolved metagenomics ,Infant gut microbiome ,community ecology ,early-life gut colonization ,Medicine (General) ,R5-920 - Abstract
Summary: Gut microbiome succession affects infant development. However, it remains unclear what factors promote persistence of initial bacterial colonizers in the developing gut. Here, we perform strain-resolved analyses to compare gut colonization of preterm and full-term infants throughout the first year of life and evaluate associations between strain persistence and strain origin as well as genetic potential. Analysis of fecal metagenomes collected from 13 full-term and 9 preterm infants reveals that infants’ initially distinct microbiomes converge by age 1 year. Approximately 11% of early colonizers, primarily Bacteroides and Bifidobacterium, persist during the first year of life, and those are more prevalent in full-term, compared with preterm infants. Examination of 17 mother-infant pairs reveals maternal gut strains are significantly more likely to persist in the infant gut than other strains. Enrichment in genes for surface adhesion, iron acquisition, and carbohydrate degradation may explain persistence of some strains through the first year of life.
- Published
- 2021
- Full Text
- View/download PDF
11. Genome-resolved metagenomics of eukaryotic populations during early colonization of premature infants and in hospital rooms
- Author
-
Matthew R. Olm, Patrick T. West, Brandon Brooks, Brian A. Firek, Robyn Baker, Michael J. Morowitz, and Jillian F. Banfield
- Subjects
Eukaryotes ,Metagenomics ,Genome-resolved metagenomics ,Hospital microbiome ,Neonatal intensive care unit ,Premature infants ,Microbial ecology ,QR100-130 - Abstract
Abstract Background Fungal infections are a significant cause of mortality and morbidity in hospitalized preterm infants, yet little is known about eukaryotic colonization of infants and of the neonatal intensive care unit as a possible source of colonizing strains. This is partly because microbiome studies often utilize bacterial 16S rRNA marker gene sequencing, a technique that is blind to eukaryotic organisms. Knowledge gaps exist regarding the phylogeny and microdiversity of eukaryotes that colonize hospitalized infants, as well as potential reservoirs of eukaryotes in the hospital room built environment. Results Genome-resolved analysis of 1174 time-series fecal metagenomes from 161 premature infants revealed fungal colonization of 10 infants. Relative abundance levels reached as high as 97% and were significantly higher in the first weeks of life (p = 0.004). When fungal colonization occurred, multiple species were present more often than expected by random chance (p = 0.008). Twenty-four metagenomic samples were analyzed from hospital rooms of six different infants. Compared to floor and surface samples, hospital sinks hosted diverse and highly variable communities containing genomically novel species, including from Diptera (fly) and Rhabditida (worm) for which genomes were assembled. With the exception of Diptera and two other organisms, zygosity of the newly assembled diploid eukaryote genomes was low. Interestingly, Malassezia and Candida species were present in both room and infant gut samples. Conclusions Increased levels of fungal co-colonization may reflect synergistic interactions or differences in infant susceptibility to fungal colonization. Discovery of eukaryotic organisms that have not been sequenced previously highlights the benefit of genome-resolved analyses, and low zygosity of assembled genomes could reflect inbreeding or strong selection imposed by room conditions.
- Published
- 2019
- Full Text
- View/download PDF
12. The developing premature infant gut microbiome is a major factor shaping the microbiome of neonatal intensive care unit rooms
- Author
-
Brandon Brooks, Matthew R. Olm, Brian A. Firek, Robyn Baker, David Geller-McGrath, Sophia R. Reimer, Karina R. Soenjoyo, Jennifer S. Yip, Dylan Dahan, Brian C. Thomas, Michael J. Morowitz, and Jillian F. Banfield
- Subjects
Infant gut ,Microbiome ,Built environment ,Neonatal intensive care unit ,Microbial ecology ,QR100-130 - Abstract
Abstract Background The neonatal intensive care unit (NICU) contains a unique cohort of patients with underdeveloped immune systems and nascent microbiome communities. Patients often spend several months in the same room, and it has been previously shown that the gut microbiomes of these infants often resemble the microbes found in the NICU. Little is known, however, about the identity, persistence, and absolute abundance of NICU room-associated bacteria over long stretches of time. Here, we couple droplet digital PCR (ddPCR), 16S rRNA gene surveys, and recently published metagenomics data from infant gut samples to infer the extent to which the NICU microbiome is shaped by its room occupants. Results Over 2832 swabs, wipes, and air samples were collected from 16 private-style NICU rooms housing very low birth weight (
- Published
- 2018
- Full Text
- View/download PDF
13. Strain-resolved analysis of hospital rooms and infants reveals overlap between the human and room microbiome
- Author
-
Brandon Brooks, Matthew R. Olm, Brian A. Firek, Robyn Baker, Brian C. Thomas, Michael J. Morowitz, and Jillian F. Banfield
- Subjects
Science - Abstract
It is thought that the hospital environment may contribute to infant microbiome development. Here, Brooks et al. present a genome-resolved metagenomic study of microbial genotypes from the infant gut and from neonatal intensive care unit rooms, showing that some strains are found in both infants and rooms.
- Published
- 2017
- Full Text
- View/download PDF
14. Genome-resolved metaproteomic characterization of preterm infant gut microbiota development reveals species-specific metabolic shifts and variabilities during early life
- Author
-
Weili Xiong, Christopher T. Brown, Michael J. Morowitz, Jillian F. Banfield, and Robert L. Hettich
- Subjects
Metaproteomics ,Human infant gut ,Shotgun proteomics ,Genome resolved ,Microbial metabolic functions ,Microbial ecology ,QR100-130 - Abstract
Abstract Background Establishment of the human gut microbiota begins at birth. This early-life microbiota development can impact host physiology during infancy and even across an entire life span. However, the functional stability and population structure of the gut microbiota during initial colonization remain poorly understood. Metaproteomics is an emerging technology for the large-scale characterization of metabolic functions in complex microbial communities (gut microbiota). Results We applied a metagenome-informed metaproteomic approach to study the temporal and inter-individual differences of metabolic functions during microbial colonization of preterm human infants’ gut. By analyzing 30 individual fecal samples, we identified up to 12,568 protein groups for each of four infants, including both human and microbial proteins. With genome-resolved matched metagenomics, proteins were confidently identified at the species/strain level. The maximum percentage of the proteome detected for the abundant organisms was ~45%. A time-dependent increase in the relative abundance of microbial versus human proteins suggested increasing microbial colonization during the first few weeks of early life. We observed remarkable variations and temporal shifts in the relative protein abundances of each organism in these preterm gut communities. Given the dissimilarity of the communities, only 81 microbial EggNOG orthologous groups and 57 human proteins were observed across all samples. These conserved microbial proteins were involved in carbohydrate, energy, amino acid and nucleotide metabolism while conserved human proteins were related to immune response and mucosal maturation. We identified seven proteome clusters for the communities and showed infant gut proteome profiles were unstable across time and not individual-specific. Applying a gut-specific metabolic module (GMM) analysis, we found that gut communities varied primarily in the contribution of nutrient (carbohydrates, lipids, and amino acids) utilization and short-chain fatty acid production. Conclusions Overall, this study reports species-specific proteome profiles and metabolic functions of human gut microbiota during early colonization. In particular, our work contributes to reveal microbiota-associated shifts and variations in the metabolism of three major nutrient sources and short-chain fatty acid during colonization of preterm infant gut.
- Published
- 2017
- Full Text
- View/download PDF
15. Candidatus Mycoplasma girerdii replicates, diversifies, and co-occurs with Trichomonas vaginalis in the oral cavity of a premature infant
- Author
-
Elizabeth K. Costello, Christine L. Sun, Erica M. Carlisle, Michael J. Morowitz, Jillian F. Banfield, and David A. Relman
- Subjects
Medicine ,Science - Abstract
Abstract Genital mycoplasmas, which can be vertically transmitted, have been implicated in preterm birth, neonatal infections, and chronic lung disease of prematurity. Our prior work uncovered 16S rRNA genes belonging to a novel, as-yet-uncultivated mycoplasma (lineage ‘Mnola’) in the oral cavity of a premature neonate. Here, we characterize the organism’s associated community, growth status, metabolic potential, and population diversity. Sequencing of genomic DNA from the infant’s saliva yielded 1.44 Gbp of high-quality, non-human read data, from which we recovered three essentially complete (including ‘Mnola’) and three partial draft genomes (including Trichomonas vaginalis). The completed 629,409-bp ‘Mnola’ genome (Candidatus Mycoplasma girerdii str. UC-B3) was distinct at the strain level from its closest relative, vaginally-derived Ca. M. girerdii str. VCU-M1, which is also associated with T. vaginalis. Replication rate measurements indicated growth of str. UC-B3 within the infant. Genes encoding surface-associated proteins and restriction-modification systems were especially diverse within and between strains. In UC-B3, the population genetic underpinnings of phase variable expression were evident in vivo. Unique among mycoplasmas, Ca. M. girerdii encodes pyruvate-ferredoxin oxidoreductase and may be sensitive to metronidazole. This study reveals a metabolically unique mycoplasma colonizing a premature neonate, and establishes the value of genome-resolved metagenomics in tracking phase variation.
- Published
- 2017
- Full Text
- View/download PDF
16. Hospitalized Premature Infants Are Colonized by Related Bacterial Strains with Distinct Proteomic Profiles
- Author
-
Christopher T. Brown, Weili Xiong, Matthew R. Olm, Brian C. Thomas, Robyn Baker, Brian Firek, Michael J. Morowitz, Robert L. Hettich, and Jillian F. Banfield
- Subjects
human microbiome ,metaproteomics ,microbial colonization ,microbial genomics ,necrotizing enterocolitis ,neonates ,Microbiology ,QR1-502 - Abstract
ABSTRACT During the first weeks of life, microbial colonization of the gut impacts human immune system maturation and other developmental processes. In premature infants, aberrant colonization has been implicated in the onset of necrotizing enterocolitis (NEC), a life-threatening intestinal disease. To study the premature infant gut colonization process, genome-resolved metagenomics was conducted on 343 fecal samples collected during the first 3 months of life from 35 premature infants housed in a neonatal intensive care unit, 14 of whom developed NEC, and metaproteomic measurements were made on 87 samples. Microbial community composition and proteomic profiles remained relatively stable on the time scale of a week, but the proteome was more variable. Although genetically similar organisms colonized many infants, most infants were colonized by distinct strains with metabolic profiles that could be distinguished using metaproteomics. Microbiome composition correlated with infant, antibiotics administration, and NEC diagnosis. Communities were found to cluster into seven primary types, and community type switched within infants, sometimes multiple times. Interestingly, some communities sampled from the same infant at subsequent time points clustered with those of other infants. In some cases, switches preceded onset of NEC; however, no species or community type could account for NEC across the majority of infants. In addition to a correlation of protein abundances with organism replication rates, we found that organism proteomes correlated with overall community composition. Thus, this genome-resolved proteomics study demonstrated that the contributions of individual organisms to microbiome development depend on microbial community context. IMPORTANCE Humans are colonized by microbes at birth, a process that is important to health and development. However, much remains to be known about the fine-scale microbial dynamics that occur during the colonization period. We conducted a genome-resolved study of microbial community composition, replication rates, and proteomes during the first 3 months of life of both healthy and sick premature infants. Infants were found to be colonized by similar microbes, but each underwent a distinct colonization trajectory. Interestingly, related microbes colonizing different infants were found to have distinct proteomes, indicating that microbiome function is not only driven by which organisms are present, but also largely depends on microbial responses to the unique set of physiological conditions in the infant gut.
- Published
- 2018
- Full Text
- View/download PDF
17. Machine Learning Leveraging Genomes from Metagenomes Identifies Influential Antibiotic Resistance Genes in the Infant Gut Microbiome
- Author
-
Sumayah F. Rahman, Matthew R. Olm, Michael J. Morowitz, and Jillian F. Banfield
- Subjects
Clostridium difficile ,antibiotic resistance ,genome-resolved metagenomics ,infant ,machine learning ,microbiome ,Microbiology ,QR1-502 - Abstract
ABSTRACT Antibiotic resistance in pathogens is extensively studied, and yet little is known about how antibiotic resistance genes of typical gut bacteria influence microbiome dynamics. Here, we leveraged genomes from metagenomes to investigate how genes of the premature infant gut resistome correspond to the ability of bacteria to survive under certain environmental and clinical conditions. We found that formula feeding impacts the resistome. Random forest models corroborated by statistical tests revealed that the gut resistome of formula-fed infants is enriched in class D beta-lactamase genes. Interestingly, Clostridium difficile strains harboring this gene are at higher abundance in formula-fed infants than C. difficile strains lacking this gene. Organisms with genes for major facilitator superfamily drug efflux pumps have higher replication rates under all conditions, even in the absence of antibiotic therapy. Using a machine learning approach, we identified genes that are predictive of an organism’s direction of change in relative abundance after administration of vancomycin and cephalosporin antibiotics. The most accurate results were obtained by reducing annotated genomic data to five principal components classified by boosted decision trees. Among the genes involved in predicting whether an organism increased in relative abundance after treatment are those that encode subclass B2 beta-lactamases and transcriptional regulators of vancomycin resistance. This demonstrates that machine learning applied to genome-resolved metagenomics data can identify key genes for survival after antibiotics treatment and predict how organisms in the gut microbiome will respond to antibiotic administration. IMPORTANCE The process of reconstructing genomes from environmental sequence data (genome-resolved metagenomics) allows unique insight into microbial systems. We apply this technique to investigate how the antibiotic resistance genes of bacteria affect their ability to flourish in the gut under various conditions. Our analysis reveals that strain-level selection in formula-fed infants drives enrichment of beta-lactamase genes in the gut resistome. Using genomes from metagenomes, we built a machine learning model to predict how organisms in the gut microbial community respond to perturbation by antibiotics. This may eventually have clinical applications.
- Published
- 2018
- Full Text
- View/download PDF
18. Strain-resolved microbial community proteomics reveals simultaneous aerobic and anaerobic function during gastrointestinal tract colonization of a preterm infant
- Author
-
Brandon eBrooks, Ryan S. Mueller, Jacque C. Young, Michael J. Morowitz, Robert L. Hettich, and Jillian F. Banfield
- Subjects
microbiome ,Metaproteomics ,microbial ecology and physiology ,colonization ,infant gut ,Microbiology ,QR1-502 - Abstract
While there has been growing interest in the gut microbiome in recent years, it remains unclear whether closely related species and strains have similar or distinct functional roles and if organisms capable of both aerobic and anaerobic growth do so simultaneously. To investigate these questions, we implemented a high-throughput mass spectrometry-based proteomics approach to identify proteins in fecal samples collected on days of life 13-21 from an infant born at 28 weeks gestation. No prior studies have coupled strain-resolved community metagenomics to proteomics for such a purpose. Sequences were manually curated to resolve the genomes of two strains of Citrobacter that were present during the later stage of colonization. Proteome extracts from fecal samples were processed via a nano-2D-LC-MS/MS and peptides were identified based on information predicted from the genome sequences for the dominant organisms, Serratia and the two Citrobacter strains. These organisms are facultative anaerobes, and proteomic information indicates the utilization of both aerobic and anaerobic metabolisms throughout the time series. This may indicate growth in distinct niches within the gastrointestinal tract. We uncovered differences in the physiology of coexisting Citrobacter strains, including differences in motility and chemotaxis functions. Additionally, for both Citrobacter strains we resolved a community-essential role in vitamin metabolism and a predominant role in propionate production. Finally, in this case study we detected differences between genome abundance and activity levels for the dominant populations. This underlines the value in layering proteomic information over genetic potential.
- Published
- 2015
- Full Text
- View/download PDF
19. Microbiome Assembly across Multiple Body Sites in Low-Birthweight Infants
- Author
-
Elizabeth K. Costello, Erica M. Carlisle, Elisabeth M. Bik, Michael J. Morowitz, and David A. Relman
- Subjects
Microbiology ,QR1-502 - Abstract
ABSTRACT The purpose of this study was to evaluate the composition and richness of bacterial communities associated with low-birthweight (LBW) infants in relation to host body site, individual, and age. Bacterial 16S rRNA genes from saliva samples, skin swabs, and stool samples collected on postnatal days 8, 10, 12, 15, 18, and 21 from six LBW (five premature) infants were amplified, pyrosequenced, and analyzed within a comparative framework that included analogous data from normal-birthweight (NBW) infants and healthy adults. We found that body site was the primary determinant of bacterial community composition in the LBW infants. However, site specificity depended on postnatal age: saliva and stool compositions diverged over time but were not significantly different until the babies were 15 days old. This divergence was primarily driven by progressive temporal turnover in the distal gut, which proceeded at a rate similar to that of age-matched NBW infants. Neonatal skin was the most adult-like in microbiota composition, while saliva and stool remained the least so. Compositional variation among infants was marked and depended on body site and age. Only the smallest, most premature infant received antibiotics during the study period; this heralded a coexpansion of Pseudomonas aeruginosa and a novel Mycoplasma sp. in the oral cavity of this vaginally delivered, intubated patient. We conclude that concurrent molecular surveillance of multiple body sites in LBW neonates reveals a delayed compositional differentiation of the oral cavity and distal gut microbiota and, in the case of one infant, an abundant, uncultivated oral Mycoplasma sp., recently detected in human vaginal samples. IMPORTANCE Complications of premature birth are the most common cause of neonatal mortality. Colonization by the indigenous microbiota, which begins at delivery, may predispose some high-risk newborns to invasive infection or necrotizing enterocolitis (NEC), and protect others, yet neonatal microbiome dynamics are poorly understood. Here, we present the first cultivation-independent time series tracking microbiota assembly across multiple body sites in a synchronous cohort of hospitalized low-birthweight (LBW) neonates. We take advantage of archived samples and publically available sequence data and compare our LBW infant findings to those from normal-birthweight (NBW) infants and healthy adults. Our results suggest potential windows of opportunity for the dispersal of microbes within and between hosts and support recent findings of substantial baseline spatiotemporal variation in microbiota composition among high-risk newborns.
- Published
- 2013
- Full Text
- View/download PDF
20. Temporal and Spatial Changes in the Microbiome Following Pediatric Severe Traumatic Brain Injury
- Author
-
Matthew B, Rogers, Dennis, Simon, Brian, Firek, Laurie, Silfies, Anthony, Fabio, Michael J, Bell, Andrew, Yeh, Justin, Azar, Richard, Cheek, Patrick M, Kochanek, Shyamal D, Peddada, and Michael J, Morowitz
- Subjects
Bacteria ,Critical Illness ,Microbiota ,RNA, Ribosomal, 16S ,Brain Injuries, Traumatic ,Pediatrics, Perinatology and Child Health ,Humans ,Child ,Critical Care and Intensive Care Medicine ,Article - Abstract
OBJECTIVE: The microbiome may be affected by trauma and critical illness. Many studies of the microbiome in critical illness are restricted to a single body site or timepoint and confounded by pre-existing conditions. We report temporal and spatial alterations in the microbiome of previously healthy children with severe traumatic brain injury (TBI). DESIGN: We collected oral, rectal, and skin swabs within 72 hours of admission and then twice weekly until ICU discharge. Samples were analyzed by 16S rRNA gene amplicon sequencing. Children undergoing elective outpatient surgery served as controls. Alpha and beta diversity comparisons were performed with Phyloseq and differentially abundant taxa were predicted using ANCOM. SETTING: Five quaternary care pediatric intensive care units PATIENTS: Patients < 18 years with severe TBI requiring placement of an intracranial pressure (ICP) monitor INTERVENTIONS: None MEASUREMENTS AND MAIN RESULTS: 327 samples were analyzed from 23 children with severe TBI and 35 controls. The community composition of initial oral (F=3.2756, R(2)=0.0535, p=0.012) and rectal (F=3.0702, R(2)=0.0649, p=0.007) samples differed between TBI and control patients. Rectal samples were depleted of commensal bacteria from Ruminococcaceae, Bacteroidaceae, and Lachnospiraceae families and enriched in Staphylococcaceae after TBI (p
- Published
- 2022
- Full Text
- View/download PDF
21. Contamination source modeling with SCRuB improves cancer phenotype prediction from microbiome data
- Author
-
George I. Austin, Heekuk Park, Yoli Meydan, Dwayne Seeram, Tanya Sezin, Yue Clare Lou, Brian A. Firek, Michael J. Morowitz, Jillian F. Banfield, Angela M. Christiano, Itsik Pe’er, Anne-Catrin Uhlemann, Liat Shenhav, and Tal Korem
- Subjects
Biomedical Engineering ,Molecular Medicine ,Bioengineering ,Applied Microbiology and Biotechnology ,Biotechnology - Published
- 2023
- Full Text
- View/download PDF
22. Using strain-resolved analysis to identify contamination in metagenomics data
- Author
-
Yue Clare Lou, Jordan Hoff, Matthew R. Olm, Jacob West-Roberts, Spencer Diamond, Brian A. Firek, Michael J. Morowitz, and Jillian F. Banfield
- Subjects
Microbiology (medical) ,Microbiology - Abstract
Background Metagenomics analyses can be negatively impacted by DNA contamination. While external sources of contamination such as DNA extraction kits have been widely reported and investigated, contamination originating within the study itself remains underreported. Results Here, we applied high-resolution strain-resolved analyses to identify contamination in two large-scale clinical metagenomics datasets. By mapping strain sharing to DNA extraction plates, we identified well-to-well contamination in both negative controls and biological samples in one dataset. Such contamination is more likely to occur among samples that are on the same or adjacent columns or rows of the extraction plate than samples that are far apart. Our strain-resolved workflow also reveals the presence of externally derived contamination, primarily in the other dataset. Overall, in both datasets, contamination is more significant in samples with lower biomass. Conclusion Our work demonstrates that genome-resolved strain tracking, with its essentially genome-wide nucleotide-level resolution, can be used to detect contamination in sequencing-based microbiome studies. Our results underscore the value of strain-specific methods to detect contamination and the critical importance of looking for contamination beyond negative and positive controls.
- Published
- 2022
- Full Text
- View/download PDF
23. Validation that human microbiome phages use alternative genetic coding with TAG stop read as Q
- Author
-
Samantha L. Peters, Adair L. Borges, Richard J. Giannone, Michael J. Morowitz, Jillian F. Banfield, and Robert L. Hettich
- Abstract
Metagenomic findings suggesting that bacteriophages (phages) can use genetic codes different from those of their host bacteria reveal a new dimension of phage-host interaction dynamics. Whereas reassignment of stop codons to code for amino acids has been predicted, there has been no proteomic validation of alternative coding in phages. In fact, one code where the stop codon TAG is reassigned to glutamine (code 15) has never been experimentally validated in any biological system. Here, we characterized stop codon reassignment in two crAss-like phages found in the human gut microbiome using LC-MS/MS-based metaproteomics. The proteome data from several phage structural proteins clearly demonstrates reassignment of the TAG stop codon to glutamine, establishing for the first time the expression of genetic code 15.One-Sentence SummaryMass spectrometry confirms protein expression of predicted alternate genetic coding in phage genomes from human microbiomes.
- Published
- 2022
- Full Text
- View/download PDF
24. Early Empiric Antibiotic Treatment Among Neonates With Congenital Heart Defects: A Brief Report of an Integrative Review
- Author
-
Jessica A. Davis, Rebecca Ho, Elizabeth K. Pace, Audrey Kim, Jiuann-Huey Lin, and Michael J. Morowitz
- Subjects
Critical Care and Intensive Care Medicine - Published
- 2022
- Full Text
- View/download PDF
25. The Pediatric Surgeon–Scientist: Succeeding in Today's Academic Environment
- Author
-
David J. Hackam, Kevin P. Mollen, Emily H. Steen, Michael J. Morowitz, Allan M. Goldstein, Ankush Gosain, Chad M. Moles, and Sundeep G. Keswani
- Subjects
Surgeons ,Surgical research ,medicine.medical_specialty ,Medical education ,Biomedical Research ,Science ,Academies and Institutes ,Pediatric Surgeon ,Pediatrics ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Pediatric surgery ,Research environment ,medicine ,Humans ,030211 gastroenterology & hepatology ,Surgery ,Psychology ,Health funding - Abstract
Background Pediatric surgeons have long been advocates of basic science research. However, new challenges facing the scientific community have threatened the success of academic surgeons pursuing basic science careers. The purpose of this study was to compare academic pediatric surgeons’ perceptions of their ability to effectively conduct basic science research to those of other surgical subspecialties. Methods An online survey was distributed to all members of the Association for Academic Surgery and Society of University Surgeons. A total of 1033 members (41%) responded, and 137 (13.3%) were pediatric surgeons. Comparisons were made between the five most-represented surgical subspecialties. Data are presented as reporting percentage and P values by Student's t-test. Results Among the specialists studied, pediatric surgeons are those most likely to believe that surgeons can succeed as basic scientists in today's research environment. Pediatric surgery reported the highest rates of National Institutes of Health funding of all surgical specialties and the lowest rates of perceived external pressures related to clinical demands, hospital administrative duties, and work–life balance concerns than their surgical peers. Conclusions Pediatric surgeons have a more optimistic perspective on the state of basic science research in surgery while exhibiting an enhanced ability to overcome the challenges that surgeon–scientists currently face. Our findings suggest that pediatric surgery may provide a model for succeeding in basic science in today's challenging surgical research environment.
- Published
- 2019
- Full Text
- View/download PDF
26. Maternal IgA protects against the development of necrotizing enterocolitis in preterm infants
- Author
-
Michael J. Morowitz, Junyi Ji, Timothy W. Hand, Kathyayini P. Gopalakrishna, Justin T. Tometich, Benjamin R Macadangdang, Ansen H.P. Burr, Robyn Baker, Misty Good, Brian Firek, Congrong Ma, and Matthew B. Rogers
- Subjects
0301 basic medicine ,IgA binding ,Immunoglobulin A ,Adult ,Male ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Enterobacteriaceae ,Pregnancy ,Enterocolitis, Necrotizing ,Medicine ,Animals ,Humans ,Microbiome ,Child ,Immunity, Mucosal ,Enterocolitis ,biology ,Host Microbial Interactions ,business.industry ,Microbiota ,Infant, Newborn ,Infant ,General Medicine ,medicine.disease ,digestive system diseases ,Mice, Inbred C57BL ,030104 developmental biology ,Mucosal immunology ,030220 oncology & carcinogenesis ,Child, Preschool ,Necrotizing enterocolitis ,Immunology ,biology.protein ,Female ,Antibody ,medicine.symptom ,business ,Infant, Premature - Abstract
Neonates are protected from colonizing bacteria by antibodies secreted into maternal milk. Necrotizing enterocolitis (NEC) is a disease of neonatal preterm infants with high morbidity and mortality that is associated with intestinal inflammation driven by the microbiota1–3. The incidence of NEC is substantially lower in infants fed with maternal milk, although the mechanisms that underlie this benefit are not clear4–6. Here we show that maternal immunoglobulin A (IgA) is an important factor for protection against NEC. Analysis of IgA binding to fecal bacteria from preterm infants indicated that maternal milk was the predominant source of IgA in the first month of life and that a relative decrease in IgA-bound bacteria is associated with the development of NEC. Sequencing of IgA-bound and unbound bacteria revealed that before the onset of disease, NEC was associated with increasing domination by Enterobacteriaceae in the IgA-unbound fraction of the microbiota. Furthermore, we confirmed that IgA is critical for preventing NEC in a mouse model, in which pups that are reared by IgA-deficient mothers are susceptible to disease despite exposure to maternal milk. Our findings show that maternal IgA shapes the host–microbiota relationship of preterm neonates and that IgA in maternal milk is a critical and necessary factor for the prevention of NEC. Immunoglobulin A antibodies in maternal milk are required for prevention of necrotizing enterocolitis in preterm neonates.
- Published
- 2019
27. Genetic and behavioral adaptation of Candida parapsilosis to the microbiome of hospitalized infants revealed by in situ genomics, transcriptomics, and proteomics
- Author
-
Michael J. Morowitz, Samantha L. Peters, Haley Gause, Robyn Baker, Feiqiao Brian Yu, Robert L. Hettich, Matthew R. Olm, Alexander D. Johnson, Yue Clare Lou, Patrick T. West, Brian Firek, and Jillian F. Banfield
- Subjects
Proteomics ,Candida parapsilosis ,Genome ,Microbial ecology ,Infant Mortality ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Aetiology ,Candida ,Genetics ,Pediatric ,0303 health sciences ,biology ,Ecology ,Premature infants ,Microbiota ,QR100-130 ,Candidiasis ,Hospital microbiome ,Infectious Diseases ,Medical Microbiology ,Proteome ,Infection ,Biotechnology ,Microbiology (medical) ,Genomics ,Microbial Sensitivity Tests ,Microbiology ,03 medical and health sciences ,Neonatal intensive care unit ,Humans ,Microbiome ,Gene ,Genome-resolved metagenomics ,030304 developmental biology ,Strain-tracking ,030306 microbiology ,Research ,Human Genome ,Infant, Newborn ,Infant ,biology.organism_classification ,Newborn ,Microbial eukaryotes ,Metagenomics ,Metaproteomics ,Transcriptome - Abstract
Background Candida parapsilosis is a common cause of invasive candidiasis, especially in newborn infants, and infections have been increasing over the past two decades. C. parapsilosis has been primarily studied in pure culture, leaving gaps in understanding of its function in a microbiome context. Results Here, we compare five unique C. parapsilosis genomes assembled from premature infant fecal samples, three of which are newly reconstructed, and analyze their genome structure, population diversity, and in situ activity relative to reference strains in pure culture. All five genomes contain hotspots of single nucleotide variants, some of which are shared by strains from multiple hospitals. A subset of environmental and hospital-derived genomes share variants within these hotspots suggesting derivation of that region from a common ancestor. Four of the newly reconstructed C. parapsilosis genomes have 4 to 16 copies of the gene RTA3, which encodes a lipid translocase and is implicated in antifungal resistance, potentially indicating adaptation to hospital antifungal use. Time course metatranscriptomics and metaproteomics on fecal samples from a premature infant with a C. parapsilosis blood infection revealed highly variable in situ expression patterns that are distinct from those of similar strains in pure cultures. For example, biofilm formation genes were relatively less expressed in situ, whereas genes linked to oxygen utilization were more highly expressed, indicative of growth in a relatively aerobic environment. In gut microbiome samples, C. parapsilosis co-existed with Enterococcus faecalis that shifted in relative abundance over time, accompanied by changes in bacterial and fungal gene expression and proteome composition. Conclusions The results reveal potentially medically relevant differences in Candida function in gut vs. laboratory environments, and constrain evolutionary processes that could contribute to hospital strain persistence and transfer into premature infant microbiomes.
- Published
- 2021
28. Dynamics of the preterm gut microbiome in health and disease
- Author
-
Michael J. Morowitz, Venkatesh Sampath, Alain Cuna, Shahid Umar, and Ishfaq Ahmed
- Subjects
0301 basic medicine ,Physiology ,Future risk ,Gestational Age ,Disease ,Bioinformatics ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,Enterocolitis, Necrotizing ,Risk Factors ,030225 pediatrics ,Physiology (medical) ,medicine ,Animals ,Humans ,Microbiome ,Hepatology ,Bacteria ,business.industry ,Probiotics ,Gastroenterology ,Infant, Newborn ,Fecal Microbiota Transplantation ,Mini-Review ,medicine.disease ,Prognosis ,Gut microbiome ,Gastrointestinal Microbiome ,Intestines ,030104 developmental biology ,Prebiotics ,Metagenomics ,Necrotizing enterocolitis ,Host-Pathogen Interactions ,Dysbiosis ,Neonatal Sepsis ,business ,Infant, Premature - 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.
- Published
- 2021
29. Time-dependent displacement of commensal skin microbes by pathogens at the site of colorectal surgery
- Author
-
David S. Medich, Jennifer Holder-Murray, Matthew B. Rogers, Michael J. Morowitz, Andrew Yeh, James P. Celebrezze, Brandon Mahler, and Brian Firek
- Subjects
Adult ,0301 basic medicine ,Microbiology (medical) ,medicine.medical_specialty ,medicine.medical_treatment ,Human skin ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,RNA, Ribosomal, 16S ,Internal medicine ,medicine ,Humans ,Surgical Wound Infection ,Microbiome ,Skin ,Colectomy ,biology ,business.industry ,Microbiota ,Perioperative ,medicine.disease ,biology.organism_classification ,Colorectal surgery ,030104 developmental biology ,Infectious Diseases ,Enterococcus ,030220 oncology & carcinogenesis ,Dysbiosis ,business ,Colorectal Surgery ,Surgical incision - Abstract
Background Although the healthy human skin microbiome has been the subject of recent studies, it is not known whether alterations among commensal microbes contribute to surgical site infections (SSIs). Our objective in this study was to characterize temporal and spatial variation in the skin microbiota of patients undergoing colorectal surgery and determine if dysbiosis contributes to SSIs. Methods Sixty one adults scheduled to undergo elective colon or rectal resection were identified by convenience sampling. By analyzing bacterial 16S rRNA gene sequences isolated from clinical samples, we used a culture-independent strategy to monitor perioperative changes in microbial diversity of fecal samples and the skin. Results A total of 990 samples from 61 patients were analyzed. Alpha diversity on the skin decreased after surgery but later recovered at the postoperative clinic visit. In most patients, we observed a transient postoperative loss of skin commensals (Corynebacterium and Propionibacterium) at the surgical site, which were replaced by potential pathogens and intestinal anaerobes (eg, Enterobacteriaceae). These changes were not observed on skin that was uninvolved in the surgical incision (chest wall). One patient developed a wound infection. Incisional skin swabs from this patient demonstrated a sharp postoperative increase in the abundance of Enterococcus, which was also cultured from wound drainage. Conclusions We observed reproducible perioperative changes in the skin microbiome following surgery. The low incidence of SSIs in this cohort precluded analysis of associations between dysbiosis and infection. We postulate that real-time monitoring of the skin microbiome could provide actionable findings about the pathogenesis of SSIs.
- Published
- 2020
- Full Text
- View/download PDF
30. Depletion of gut microbiota is associated with improved neurologic outcome following traumatic brain injury
- Author
-
Keri Janesko-Feldman, Vincent Vagni, Robert S. B. Clark, Yuan Gao, Dennis W. Simon, Matthew B. Rogers, Michael J. Morowitz, Kevin P. Mollen, Garret Vincent, John A. Ozolek, Patrick M. Kochanek, and Brian Firek
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Traumatic brain injury ,Gut–brain axis ,Inflammation ,Gut flora ,Hippocampal formation ,Gastroenterology ,Hippocampus ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Brain Injuries, Traumatic ,medicine ,Animals ,Fear conditioning ,Molecular Biology ,Neurons ,biology ,business.industry ,General Neuroscience ,Head injury ,Neomycin ,Recovery of Function ,medicine.disease ,biology.organism_classification ,Gastrointestinal Microbiome ,Disease Models, Animal ,030104 developmental biology ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Developmental Biology ,medicine.drug - Abstract
Signaling between intestinal microbiota and the brain influences neurologic outcome in multiple forms of brain injury. The impact of gut microbiota following traumatic brain injury (TBI) has not been well established. Our objective was to compare TBI outcomes in specific pathogen-free mice with or without depletion of intestinal bacteria. Adult male C57BL6/J SPF mice (n=6/group) were randomized to standard drinking water or ampicillin (1g/L), metronidazole (1g/L), neomycin (1g/L), and vancomycin (0.5g/L) (AMNV) containing drinking water 14 days prior to controlled cortical impact (CCI) model of TBI. 16S rRNA gene sequencing of fecal pellets was performed and alpha and beta diversity determined. Hippocampal neuronal density and microglial activation was assessed 72 hours post-injury by immunohistochemistry. In addition, mice (n=8–12/group) were randomized to AMNV or no treatment initiated immediately after CCI and memory acquisition (fear conditioning) and lesion volume assessed. Mice receiving AMNV had significantly reduced alpha diversity (p
- Published
- 2020
31. Use of C-Reactive Protein and Ferritin Biomarkers in Daily Pediatric Practice
- Author
-
Matthew D. Taylor, Michael J. Morowitz, Kathryn Torok, Michael L. Moritz, Andrew J. Nowalk, Vivek Allada, Rakesh Sindhi, Rajesh Aneja, Joseph A. Carcillo, and Marian G. Michaels
- Subjects
medicine.medical_specialty ,Clinical Decision-Making ,MEDLINE ,Disease ,Infections ,Significant elevation ,Pediatrics ,Article ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Reference Values ,030225 pediatrics ,Clinical Decision Rules ,medicine ,Humans ,030212 general & internal medicine ,Intensive care medicine ,Child ,Inflammation ,Pediatric practice ,biology ,business.industry ,C-reactive protein ,Clinical Practice ,Ferritin ,Clinical research ,C-Reactive Protein ,Pediatrics, Perinatology and Child Health ,Ferritins ,biology.protein ,business ,Biomarkers - Abstract
PRACTICE GAP: Recent reports have detailed the use of these widely available biomarkers in diagnosing and predicting outcomes in a wide array of clinical diseases, most of which are severe, life-threatening, and difficult to diagnose. Many pediatricians have difficulty understanding and using the results of C-reactive protein and ferritin blood tests, which are easily accessible in clinical practice. This summary of the literature can assist in the interpretation and application of these useful and readily available tools in their clinical practice. BACKGROUND: Recent pediatric clinical research has begun to focus on risk stratification tools using multibiomarker models. C-reactive protein (CRP) and ferritin biomarkers are widely available and used to varying degrees in daily practice, but there is no single source examining the evidence behind their use. OBJECTIVES: To summarize the evidence behind the use of CRP and ferritin biomarkers in pediatric practice and to begin development of a consensus for their future use for pediatricians. EVIDENCE: All the literature involving CRP and ferritin in pediatrics available on PubMed was surveyed. Research applicable to daily pediatric practice was summarized in the body of the article. Pediatric clinicians of various subspecialties contributed to the summary of the use of CRP and ferritin biomarkers in clinical practice in various disease processes. A clinical decision pathway is described, and evidence is summarized. FINDINGS: CRP and ferritin biomarkers have diverse uses with various cutoff values in the literature, making their use in daily practice difficult. Elevation of these markers coincides with their significant elevation in uncontrolled inflammation. CONCLUSIONS: CRP and ferritin biomarkers are widely used in pediatrics. This review provides a resource summarizing evidence into a single source. There is sufficient evidence to indicate that these biomarkers of inflammation can be useful in guiding clinical decision making in specific clinical scenarios; however, further work is needed to improve their use in clinical practice.
- Published
- 2020
32. Genetic and behavioral adaptation of Candida parapsilosis to the microbiome of hospitalized infants revealed by in situ genomics, transcriptomics and proteomics
- Author
-
Robert L. Hettich, Samantha L. Peters, Matthew R. Olm, Brian Firek, Feiqiao Brian Yu, Alexander D. Johnson, Jillian F. Banfield, Robyn Baker, Patrick T. West, Yue Clare Lou, and Michael J. Morowitz
- Subjects
Genetics ,0303 health sciences ,biology ,030306 microbiology ,Genomics ,Context (language use) ,Candida parapsilosis ,biology.organism_classification ,Genome ,3. Good health ,03 medical and health sciences ,Proteome ,Metaproteomics ,Microbiome ,Gene ,030304 developmental biology - Abstract
Candida parapsilosisis a common cause of invasive candidiasis, especially in newborn infants, and infections have been increasing over the past two decades.C. parapsilosishas been primarily studied in pure culture, leaving gaps in understanding of its function in microbiome context. Here, we reconstructed five uniqueC. parapsilosisgenomes from premature infant fecal samples and analyzed their genome structure, population diversity andin situactivity relative to reference strains in pure culture. All five genomes contain hotspots of single nucleotide variants, some of which are shared by strains from multiple hospitals. A subset of environmental and hospital-derived genomes share variants within these hotspots suggesting derivation of that region from a common ancestor. Four of the newly reconstructedC. parapsilosisgenomes have four to sixteen copies of the gene RTA3, which encodes a lipid translocase and is implicated in antifungal resistance, potentially indicating adaptation to hospital antifungal use. Time course metatranscriptomics and metaproteomics on fecal samples from a premature infant with aC. parapsilosisblood infection revealed highly variablein situexpression patterns that are distinct from those of similar strains in pure cultures. For example, biofilm formation genes were relatively less expressedin situ, whereas genes linked to oxygen utilization were more highly expressed, indicative of growth in a relatively aerobic environment. In gut microbiome samples,C. parapsilosiscoexisted withEnterococcus faecalisthat shifted in relative abundance over time, accompanied by changes in bacterial and fungal gene expression and proteome composition. The results reveal potentially medically relevant differences in Candida function in gut vs. laboratory environments, and constrain evolutionary processes that could contribute to hospital strain persistence and transfer into premature infant microbiomes.
- Published
- 2020
- Full Text
- View/download PDF
33. Probiotics and fecal microbiota transplantation in surgical disorders
- Author
-
Michael J. Morowitz and Andrew Yeh
- Subjects
0301 basic medicine ,biology ,Colorectal cancer ,business.industry ,Gastroenterology ,Disease ,Gut flora ,medicine.disease ,biology.organism_classification ,Bioinformatics ,Inflammatory bowel disease ,law.invention ,Clostridium Difficile Colitis ,03 medical and health sciences ,Probiotic ,030104 developmental biology ,0302 clinical medicine ,Immune system ,law ,Immunology ,medicine ,030211 gastroenterology & hepatology ,Surgery ,Microbiome ,business - Abstract
The importance of the gut microbiota in health and disease has led to interest in developing methods to modify it. Probiotics administration and fecal microbiota transplantation (FMT) are two such approaches that can alter the gut microbiota, potentially offering health benefits by blocking gut colonization by pathogenic organisms and preventing a maladaptive immune response. Both methods have been studied in a variety of settings relevant to colorectal surgeons, including colorectal cancer, inflammatory bowel disease, Clostridium difficile colitis, and surgical site infections. However, both therapies offer risks and benefits in surgical patients. Probiotics allow for targeted alterations of the microbiome, but lingering questions remain regarding strain selection. FMT offers to more completely restore the healthy gut microbial ecosystem but it is difficult to study in animals and to determine its precise mechanism of action. Standardizing study methodologies and using modern molecular and genetic techniques to elucidate the mechanisms of action will be needed to determine the role of probiotic administration and FMT in treating or preventing complications in patients undergoing major abdominal surgery.
- Published
- 2018
- Full Text
- View/download PDF
34. Rapid Single-Cell Microbiological Analysis: Toward Precision Management of Infections and Dysbiosis
- Author
-
Pak Kin Wong, Hui Li, Michael J. Morowitz, and Neal J. Thomas
- Subjects
Automation, Laboratory ,medicine.medical_specialty ,Miniaturization ,Bacteria ,business.industry ,Microfluidics ,Bacterial Infections ,medicine.disease ,humanities ,Computer Science Applications ,Medical Laboratory Technology ,PNAS Plus ,Point-of-Care Testing ,Lab-On-A-Chip Devices ,medicine ,Dysbiosis ,Humans ,Single-Cell Analysis ,Intensive care medicine ,business ,health care economics and organizations - Abstract
Bacterial infection is a leading cause of morbidity and mortality (from infants to the elderly) and accounts for more than $20 billion in healthcare costs in the United States each year. The pathogens responsible for many of the common infectious diseases, such as urinary tract infection (UTI) and ventilator-associated infections (VAIs), have proven to be highly adept in acquiring mechanisms of antimicrobial resistance. The use of broad-spectrum antibiotics by healthcare providers and the infiltration of antibiotics in the environment have accelerated the selection and growth of resistant pathogens. To further exacerbate the problem, the need for new antibiotics has far outpaced the development of new classes of antibiotics by the pharmaceutical industry (only two new classes of antibiotics have reached the market in the last 20 years), in large part due to prohibitive cost and historically poor return on investment to develop new antibiotics. Consequently, clinicians have limited treatment options, particularly in the neediest patients. To tackle this major global health issue, we are developing novel technological approaches for rapid definitive clinical microbiological analysis. These technologies will improve the clinical management of bacterial infections and reduce the improper use of antibiotics in current practice, hopefully limiting the spread of drug-resistant organisms.
- Published
- 2019
- Full Text
- View/download PDF
35. Safety, Clinical Response, and Microbiome Findings Following Fecal Microbiota Transplant in Children With Inflammatory Bowel Disease
- Author
-
Alka Goyal, Leah Siebold, Adam Kufen, Brian Firek, Matthew B. Rogers, Brian R. Bush, Michael J. Morowitz, and Andrew Yeh
- Subjects
0301 basic medicine ,medicine.medical_specialty ,biology ,business.industry ,Gastroenterology ,Disease ,medicine.disease ,biology.organism_classification ,Ulcerative colitis ,Inflammatory bowel disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Refractory ,Fusobacterium ,Internal medicine ,Haemophilus ,medicine ,Immunology and Allergy ,030211 gastroenterology & hepatology ,Microbiome ,Adverse effect ,business - Abstract
Background The role of fecal microbiota transplant (FMT) in the treatment of pediatric inflammatory bowel disease (IBD) is unknown. The aims of this study were to assess safety, clinical response, and gut microbiome alterations in children with Crohn's disease (CD), ulcerative colitis (UC), or indeterminate colitis (IC). Methods In this open-label, single-center prospective trial, patients with IBD refractory to medical therapy underwent a single FMT by upper and lower endoscopy. Adverse events, clinical response, gut microbiome, and biomarkers were assessed at baseline, 1 week, 1 month, and 6 months following FMT. Results Twenty-one subjects were analyzed, with a median age of 12 years, of whom 57% and 28% demonstrated clinical response at 1 and 6 months post-FMT, respectively. Two CD patients were in remission at 6 months. Adverse events attributable to FMT were mild to moderate and self-limited. Patients prior to FMT showed decreased species diversity and significant microbiome compositional differences characterized by increased Enterobacteriaceae, Enterococcus, Haemophilus, and Fusobacterium compared with donors and demonstrated increased species diversity at 30 days post-FMT. At 6 months, these changes shifted toward baseline. Clinical responders had a higher relative abundance of Fusobacterium and a lower diversity at baseline, as well as a greater shift toward donor-like microbiome after FMT compared with nonresponders. Conclusions A single FMT is relatively safe and can result in a short-term response in young patients with active IBD. Responders possessed increased Fusobacterium prior to FMT and demonstrated more significant microbiome changes compared with nonresponders after FMT. Microbiome characteristics may help in predicting response.
- Published
- 2018
- Full Text
- View/download PDF
36. Candidatus Mycoplasma girerdii replicates, diversifies, and co-occurs with Trichomonas vaginalis in the oral cavity of a premature infant
- Author
-
Christine L. Sun, Jillian F. Banfield, Elizabeth K. Costello, David A. Relman, Erica M. Carlisle, and Michael J. Morowitz
- Subjects
0301 basic medicine ,Male ,Science ,030106 microbiology ,Population ,Trichomonas Infections ,Biology ,medicine.disease_cause ,Low Birth Weight and Health of the Newborn ,Genome ,Article ,Microbiology ,03 medical and health sciences ,Mycoplasma ,Preterm ,Infant Mortality ,medicine ,Genetics ,Trichomonas vaginalis ,Humans ,Mycoplasma Infections ,education ,Gene ,Phase variation ,Pediatric ,education.field_of_study ,Mouth ,Multidisciplinary ,Human Genome ,Infant, Newborn ,Infant ,Perinatal Period - Conditions Originating in Perinatal Period ,Newborn ,3. Good health ,Other Physical Sciences ,genomic DNA ,Good Health and Well Being ,030104 developmental biology ,Candidatus ,Medicine ,Female ,Biochemistry and Cell Biology ,Biotechnology - Abstract
Genital mycoplasmas, which can be vertically transmitted, have been implicated in preterm birth, neonatal infections, and chronic lung disease of prematurity. Our prior work uncovered 16S rRNA genes belonging to a novel, as-yet-uncultivated mycoplasma (lineage ‘Mnola’) in the oral cavity of a premature neonate. Here, we characterize the organism’s associated community, growth status, metabolic potential, and population diversity. Sequencing of genomic DNA from the infant’s saliva yielded 1.44 Gbp of high-quality, non-human read data, from which we recovered three essentially complete (including ‘Mnola’) and three partial draft genomes (including Trichomonas vaginalis). The completed 629,409-bp ‘Mnola’ genome (Candidatus Mycoplasma girerdii str. UC-B3) was distinct at the strain level from its closest relative, vaginally-derived Ca. M. girerdii str. VCU-M1, which is also associated with T. vaginalis. Replication rate measurements indicated growth of str. UC-B3 within the infant. Genes encoding surface-associated proteins and restriction-modification systems were especially diverse within and between strains. In UC-B3, the population genetic underpinnings of phase variable expression were evident in vivo. Unique among mycoplasmas, Ca. M. girerdii encodes pyruvate-ferredoxin oxidoreductase and may be sensitive to metronidazole. This study reveals a metabolically unique mycoplasma colonizing a premature neonate, and establishes the value of genome-resolved metagenomics in tracking phase variation.
- Published
- 2017
- Full Text
- View/download PDF
37. The Future of Basic Science in Academic Surgery
- Author
-
Lily S. Cheng, Herbert J. Zeh, Allan M. Goldstein, Sundeep G. Keswani, Matthew H. Levine, John S. Kuo, David J. Hackam, Nita Ahuja, Chad M. Moles, and Michael J. Morowitz
- Subjects
medicine.medical_specialty ,business.industry ,Basic science ,Background data ,ComputingMethodologies_IMAGEPROCESSINGANDCOMPUTERVISION ,ComputerApplications_COMPUTERSINOTHERSYSTEMS ,Translational research ,030230 surgery ,Surgeon scientist ,Article ,humanities ,Surgery ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,business ,health care economics and organizations - Abstract
Objective:The aim of this study was to examine the challenges confronting surgeons performing basic science research in today's academic surgery environment.Summary of Background Data:Multiple studies have identified challenges confronting surgeon-scientists and impacting their ability to be success
- Published
- 2017
- Full Text
- View/download PDF
38. Dietary Supplementation With Nonfermentable Fiber Alters the Gut Microbiota and Confers Protection in Murine Models of Sepsis
- Author
-
Valentina Di Caro, Brian Firek, Michael J. Morowitz, Matthew B. Rogers, Diana Pang, Rajesh Aneja, Jessica Cummings, Sarangarajan Ranganathan, and Robert B. Clark
- Subjects
Dietary Fiber ,0301 basic medicine ,Neutrophils ,medicine.drug_class ,Antibiotics ,Inflammation ,Gut flora ,Critical Care and Intensive Care Medicine ,Systemic inflammation ,Article ,Sepsis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,RNA, Ribosomal, 16S ,medicine ,Animals ,Prospective Studies ,030212 general & internal medicine ,Feces ,biology ,business.industry ,Lachnospiraceae ,Akkermansia ,biology.organism_classification ,medicine.disease ,Survival Analysis ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Dietary Supplements ,Immunology ,Inflammation Mediators ,medicine.symptom ,business ,Biomarkers - Abstract
Objectives Links between microbial alterations and systemic inflammation have been demonstrated in chronic disease, but little is known about these interactions during acute inflammation. This study investigates the effect of dietary supplementation with cellulose, a nonfermentable fiber, on the gut microbiota, inflammatory markers, and survival in two murine models of sepsis. Design Prospective experimental study. Setting University laboratory. Subjects Six-week-old male C57BL/6 wild-type mice. Interventions Mice were assigned to low-fiber, normal-fiber, or high-fiber diets with or without antibiotics for 2 weeks and then subjected to sepsis by cecal ligation and puncture or endotoxin injection. Fecal samples were collected for microbiota analyses before and after dietary interventions. Measurements and main results Mice that received a high-fiber diet demonstrated increased survival after cecal ligation and puncture relative to mice receiving low-fiber or normal-fiber diets. The survival benefit was associated with decreased serum concentration of pro-inflammatory cytokines, reduced neutrophil infiltration in the lungs, and diminished hepatic inflammation. The high-fiber diet also increased survival after endotoxin injection. Bacterial 16S ribosomal RNA gene sequences from each sample were amplified, sequenced, and analyzed. Fiber supplementation yielded an increase in relative abundance of the genera Akkermansia and Lachnospiraceae, taxa commonly associated with metabolic health. Administration of antibiotics to mice on the high-fiber diet negated the enrichment of Akkermansia species and the survival benefit after cecal ligation and puncture. Conclusion Dietary supplementation with cellulose offers a microbe-mediated survival advantage in murine models of sepsis. Improved understanding of the link between diet, the microbiota, and systemic illness may yield new therapeutic strategies for patients with sepsis.
- Published
- 2017
- Full Text
- View/download PDF
39. Dysbiosis in the intensive care unit: Microbiome science coming to the bedside
- Author
-
Bryan J. McVerry, Gary B. Huffnagle, Robert P. Dickson, Georgios D Kitsios, Alison Morris, and Michael J. Morowitz
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Critical Care ,Critical Illness ,Disease ,Critical Care and Intensive Care Medicine ,Article ,law.invention ,Terminology ,03 medical and health sciences ,0302 clinical medicine ,law ,Intensive care ,medicine ,Animals ,Humans ,030212 general & internal medicine ,Microbiome ,Intensive care medicine ,Evidence-Based Medicine ,business.industry ,Microbiota ,Evidence-based medicine ,medicine.disease ,Intensive care unit ,Intensive Care Units ,030104 developmental biology ,Models, Animal ,Critical illness ,Dysbiosis ,business - Abstract
Complex microbial communities within the human body, constituting the microbiome, have a broad impact on human health and disease. A growing body of research now examines the role of the microbiome in patients with critical illness, such as sepsis and acute respiratory failure. In this article, we provide an introduction to microbiome concepts and terminology and we systematically review the current evidence base of the critical-illness microbiome, including 51 studies in animal models and pediatric and adult critically-ill patients. We further examine how this emerging scientific discipline may transform the way we manage infectious and inflammatory diseases in intensive care units. The evolving molecular, culture-independent techniques offer the ability to study microbial communities in unprecedented depth and detail, and in the short-term, may enable us to diagnose and treat infections in critical care more precisely and effectively. Longer-term, these tools may also give us insights in the underlying pathophysiology of critical illness and reveal previously unsuspected targets for innovative, microbiome-targeted therapeutics. We finally propose a roadmap for future studies in the field for transforming critical care from its current isolated focus on the host to a more personalized paradigm addressing both human and microbial contributions to critical illness.
- Published
- 2017
- Full Text
- View/download PDF
40. InStrain enables population genomic analysis from metagenomic data and rigorous detection of identical microbial strains
- Author
-
Michael J. Morowitz, Matthew R. Olm, Brian Firek, Alexander Crits-Christoph, Jillian F. Banfield, and Keith Bouma-Gregson
- Subjects
Genetics ,0303 health sciences ,education.field_of_study ,Klebsiella ,Genetic diversity ,Strain (biology) ,Population ,Biology ,biology.organism_classification ,Genome ,Nucleotide diversity ,03 medical and health sciences ,0302 clinical medicine ,Metagenomics ,Microbiome ,education ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Coexisting microbial cells of the same species often exhibit genetic differences that can affect phenotypes ranging from nutrient preference to pathogenicity. Here we present inStrain, a program that utilizes metagenomic paired reads to profile intra-population genetic diversity (microdiversity) across whole genomes and compare populations in a microdiversity-aware manner, dramatically increasing genomic comparison accuracy when benchmarked against existing methods. We use inStrain to profile >1,000 fecal metagenomes from newborn premature infants and find that siblings share significantly more strains than unrelated infants, although identical twins share no more strains than fraternal siblings. Infants born via cesarean section harbored Klebsiella with significantly higher nucleotide diversity than infants delivered vaginally, potentially reflecting acquisition from hospital versus maternal microbiomes. Genomic loci showing diversity within an infant included variants found in other infants, possibly reflecting inoculation from diverse hospital-associated sources. InStrain can be applied to any metagenomic dataset for microdiversity analysis and rigorous strain comparison.
- Published
- 2020
- Full Text
- View/download PDF
41. Combined analysis of microbial metagenomic and metatranscriptomic sequencing data to assess in situ physiological conditions in the premature infant gut
- Author
-
Matthew R. Olm, Brian Firek, Christopher T. Brown, Tali Raveh-Sadka, Yonatan Sher, Jillian F. Banfield, Michael J. Morowitz, Ruth Sher, Robyn Baker, and Zoetendal, Erwin G
- Subjects
0301 basic medicine ,Male ,Molecular biology ,Low Birth Weight and Health of the Newborn ,Biochemistry ,Genome ,Transcriptome ,Database and Informatics Methods ,Sequencing techniques ,Databases, Genetic ,Infant Mortality ,Genetics ,Pediatric ,Multidisciplinary ,Microbiota ,Bacterial ,Neurochemistry ,RNA sequencing ,Genomics ,Genomic Databases ,Chemistry ,Medical Microbiology ,Physical Sciences ,Necrotizing enterocolitis ,Medicine ,Female ,Neurochemicals ,Infant, Premature ,Research Article ,Chemical Elements ,Biotechnology ,Escherichia ,General Science & Technology ,Science ,DNA transcription ,030106 microbiology ,Microbial Genomics ,Biology ,Nitric Oxide ,Research and Analysis Methods ,Microbiology ,Electron Transport Complex IV ,03 medical and health sciences ,Databases ,Rare Diseases ,Enterobacteriaceae ,Gene mapping ,Genetic ,Enterocolitis, Necrotizing ,Digestive System Physiological Phenomena ,Preterm ,medicine ,Humans ,Microbiome ,Gene ,Premature ,Bacteria ,Enterocolitis ,Gut Bacteria ,Gene Mapping ,Infant, Newborn ,Organisms ,Biology and Life Sciences ,Computational Biology ,RNA ,Infant ,Perinatal Period - Conditions Originating in Perinatal Period ,Genome Analysis ,medicine.disease ,Newborn ,Gastrointestinal Microbiome ,Oxygen ,Biological Databases ,Molecular biology techniques ,030104 developmental biology ,Good Health and Well Being ,Genes ,Genes, Bacterial ,Metagenomics ,Case-Control Studies ,Metagenome ,Gene expression ,Necrotizing ,Digestive Diseases ,Digestive System ,Neuroscience - Abstract
Microbes alter their transcriptomic profiles in response to the environment. The physiological conditions experienced by a microbial community can thus be inferred using meta-transcriptomic sequencing by comparing transcription levels of specifically chosen genes. However, this analysis requires accurate reference genomes to identify the specific genes from which RNA reads originate. In addition, such an analysis should avoid biases in transcript counts related to differences in organism abundance. In this study we describe an approach to address these difficulties. Sample-specific meta-genomic assembled genomes (MAGs) were used as reference genomes to accurately identify the origin of RNA reads, and transcript ratios of genes with opposite transcription responses were compared to eliminate biases related to differences in organismal abundance, an approach hereafter named the “diametric ratio” method. We used this approach to probe the environmental conditions experienced by Escherichia spp. in the gut of 4 premature infants, 2 of whom developed necrotizing enterocolitis (NEC), a severe inflammatory intestinal disease. We analyzed twenty fecal samples taken from four premature infants (4–6 time points from each infant), and found significantly higher diametric ratios of genes associated with low oxygen levels in samples of infants later diagnosed with NEC than in samples without NEC. We also show this method can be used for examining other physiological conditions, such as exposure to nitric oxide and osmotic pressure. These study results should be treated with caution, due to the presence of confounding factors that might also distinguish between NEC and control infants. Nevertheless, together with benchmarking analyses, we show here that the diametric ratio approach can be applied for evaluating the physiological conditions experienced by microbes in situ. Results from similar studies can be further applied for designing diagnostic methods to detect NEC in its early developmental stages.
- Published
- 2020
42. Necrotizing enterocolitis is preceded by increased gut bacterial replication, Klebsiella, and fimbriae-encoding bacteria
- Author
-
Robyn Baker, Nicholas Bhattacharya, Alexander Crits-Christoph, Michael J. Morowitz, Matthew R. Olm, Brian Firek, Yun S. Song, and Jillian F. Banfield
- Subjects
Klebsiella ,Fimbria ,Diseases and Disorders ,Diseases ,Infant, Premature, Diseases ,Low Birth Weight and Health of the Newborn ,Feces ,0302 clinical medicine ,Infant Mortality ,2.2 Factors relating to the physical environment ,Aetiology ,Research Articles ,Enterocolitis ,Pediatric ,0303 health sciences ,Multidisciplinary ,biology ,Bacterial ,SciAdv r-articles ,Enterobacteriaceae ,3. Good health ,Multigene Family ,Necrotizing enterocolitis ,medicine.symptom ,Infection ,Infant, Premature ,Research Article ,Bacterial genetics ,Microbiology ,Fimbriae ,03 medical and health sciences ,Rare Diseases ,Enterocolitis, Necrotizing ,Preterm ,Clinical Research ,medicine ,Genetics ,Humans ,Health and Medicine ,Premature ,030304 developmental biology ,Prevention ,Infant, Newborn ,Infant ,Perinatal Period - Conditions Originating in Perinatal Period ,biology.organism_classification ,medicine.disease ,Newborn ,digestive system diseases ,Gastrointestinal Microbiome ,Quorum sensing ,Orphan Drug ,Good Health and Well Being ,Fimbriae, Bacterial ,Metagenomics ,Necrotizing ,Digestive Diseases ,030217 neurology & neurosurgery ,Bacteria - Abstract
Metagenomic analysis identifies microbial signatures preceding necrotizing enterocolitis development in premature infants., Necrotizing enterocolitis (NEC) is a devastating intestinal disease that occurs primarily in premature infants. We performed genome-resolved metagenomic analysis of 1163 fecal samples from premature infants to identify microbial features predictive of NEC. Features considered include genes, bacterial strain types, eukaryotes, bacteriophages, plasmids, and growth rates. A machine learning classifier found that samples collected before NEC diagnosis harbored significantly more Klebsiella, bacteria encoding fimbriae, and bacteria encoding secondary metabolite gene clusters related to quorum sensing and bacteriocin production. Notably, replication rates of all bacteria, especially Enterobacteriaceae, were significantly higher 2 days before NEC diagnosis. The findings uncover biomarkers that could lead to early detection of NEC and targets for microbiome-based therapeutics.
- Published
- 2019
43. Clades of huge phage from across Earth’s ecosystems
- Author
-
Lesley A. Warren, Jennifer A. Doudna, Keith Bouma-Gregson, Rohan Sachdeva, Anne-Catherine Lehours, Christine He, Jamie H. D. Cate, Jinglie Zhou, Ray Keren, Alex D. Thomas, Fred R. Ward, Patrick Munk, Joanne M. Santini, Mary E. Power, Susan T.L. Harrison, Cindy J. Castelle, Alexander J. Probst, Raphaël Méheust, Christine L. Sun, Kari M. Finstad, Adi Lavy, Kelly C. Wrighton, Mikayla A. Borton, Lin-Xing Chen, Jillian F. Banfield, Karthik Anantharaman, Yuki Amano, Brandon Brooks, Ibrahim F. Farag, Daniela S. Aliaga Goltsman, David A. Relman, Ronald Amundson, Alexander L. Jaffe, Paula Matheus-Carnevali, Matthew R. Olm, Wen-Jun Li, Shufei Lei, Audra E. Devoto, Susannah G. Tringe, Tara Colenbrander Nelson, Basem Al-Shayeb, Michael J. Morowitz, and Rose S. Kantor
- Subjects
0303 health sciences ,TRNA modification ,030306 microbiology ,viruses ,Translation (biology) ,15. Life on land ,Biology ,Genome ,03 medical and health sciences ,chemistry.chemical_compound ,Eukaryotic translation ,chemistry ,Ribosomal protein ,Evolutionary biology ,Transfer RNA ,Gene ,DNA ,030304 developmental biology - Abstract
Phage typically have small genomes and depend on their bacterial hosts for replication. DNA sequenced from many diverse ecosystems revealed hundreds of huge phage genomes, between 200 kbp and 716 kbp in length. Thirty-four genomes were manually curated to completion, including the largest phage genomes yet reported. Expanded genetic repertoires include diverse and new CRISPR-Cas systems, tRNAs, tRNA synthetases, tRNA modification enzymes, translation initiation and elongation factors, and ribosomal proteins. Phage CRISPR-Cas systems have the capacity to silence host transcription factors and translational genes, potentially as part of a larger interaction network that intercepts translation to redirect biosynthesis to phage-encoded functions. In addition, some phage may repurpose bacterial CRISPR-Cas systems to eliminate competing phage. We phylogenetically define major clades of huge phage from human and other animal microbiomes, oceans, lakes, sediments, soils and the built environment. We conclude that their large gene inventories reflect a conserved biological strategy, observed over a broad bacterial host range and across Earth’s ecosystems.
- Published
- 2019
- Full Text
- View/download PDF
44. Author's Reply to Drs. Franco Scaldaferri and Valentina Petito, PhD
- Author
-
Alka Goyal and Michael J. Morowitz
- Subjects
business.industry ,Microbiota ,Gastroenterology ,Immunology and Allergy ,Medicine ,Humans ,Fecal Microbiota Transplantation ,business ,Child ,Inflammatory Bowel Diseases - Published
- 2019
45. Necrotizing enterocolitis is preceded by increased gut bacterial replication, Klebsiella, and fimbriae-encoding bacteria that may stimulate TLR4 receptors
- Author
-
Michael J. Morowitz, Nicholas Bhattacharya, Robyn Baker, Brian Firek, Yun S. Song, Alexander Crits-Christoph, Matthew R. Olm, and Jillian F. Banfield
- Subjects
0303 health sciences ,Klebsiella ,biology ,030306 microbiology ,Fimbria ,biology.organism_classification ,medicine.disease ,Enterobacteriaceae ,digestive system diseases ,3. Good health ,Microbiology ,03 medical and health sciences ,Quorum sensing ,Plasmid ,Necrotizing enterocolitis ,medicine ,Microbiome ,Bacteria ,030304 developmental biology - Abstract
Necrotizing enterocolitis (NEC) is a devastating intestinal disease that occurs primarily in premature infants. We performed genome-resolved metagenomic analysis of 1,163 fecal samples from premature infants to identify microbial features predictive of NEC. Features considered include genes, bacterial strain types, eukaryotes, bacteriophages, plasmids and growth rates. A machine learning classifier found that samples collected prior to NEC diagnosis harbored significantly more Klebsiella, bacteria encoding fimbriae, and bacteria encoding secondary metabolite gene clusters related to quorum sensing and bacteriocin production. Notably, replication rates of all bacteria, especially Enterobacteriaceae, were significantly higher two days before NEC diagnosis. The findings uncover biomarkers that could lead to early detection of NEC and targets for microbiome-based therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
46. Dysbiosis Across Multiple Body Sites in Critically Ill Adult Surgical Patients
- Author
-
Michael J. Morowitz, Matthew B. Rogers, Matthew D. Neal, Andrew Yeh, Brian Firek, and Brian S. Zuckerbraun
- Subjects
Adult ,0301 basic medicine ,medicine.medical_specialty ,Critical Illness ,Staphylococcus ,Corynebacterium ,Biology ,Critical Care and Intensive Care Medicine ,medicine.disease_cause ,Polymerase Chain Reaction ,law.invention ,Feces ,03 medical and health sciences ,Mycoplasma ,0302 clinical medicine ,Tongue ,law ,RNA, Ribosomal, 16S ,Internal medicine ,medicine ,Humans ,Microbiome ,Intensive care medicine ,Faecalibacterium ,Skin ,Microbiota ,Human microbiome ,030208 emergency & critical care medicine ,medicine.disease ,biology.organism_classification ,Intensive care unit ,Intensive Care Units ,030104 developmental biology ,Enterococcus ,Emergency Medicine ,Dysbiosis - Abstract
The microbiota of critically ill patients likely undergoes dramatic changes but has not been rigorously studied with a culture-independent high-throughput approach. The aim of this study was to characterize spatial and temporal variation in the microbiota of critically ill patients. Trauma and acute surgery patients admitted to the intensive care unit (ICU) were sampled at five body sites (stool, tongue, skin, trachea, urine) every 3 to 4 days. A mean of 10.8 samples was collected from 32 patients with a mean sampling period of 8.8 days. Bacterial 16S rRNA sequences were amplified and sequenced for microbiota analyses. Results were compared to data from unhospitalized adult participants in the American Gut and Human Microbiome Projects. Relative to healthy adults, alpha diversity was decreased in ICU gut and skin samples at all time points. Diversity in tongue swabs decreased over time. Beta diversity measures indicated differences in community membership between critically ill and healthy adults at each body site. Taxonomic alterations in the ICU included depletion of important commensal bacteria such as Faecalibacterium in GI samples and Corynebacterium in skin swabs and enrichment with pathogens such as Enterococcus, Mycoplasma, and Staphylococcus. A high proportion of ICU sample sets contained pathogens present simultaneously at three body sites indicating widespread colonization. In several cases, clinically relevant airway infections were preceded by the appearance of the causative pathogen in tracheal microbiome profiles. These results demonstrate that the microbiome of critically ill patients undergoes a loss of diversity, loss of site specificity, and a shift toward dominant pathogens. These changes may provide opportunities to precisely modulate the microbiome and thereby improve patient outcomes.
- Published
- 2016
- Full Text
- View/download PDF
47. Pilot study of sources and concentrations of size-resolved airborne particles in a neonatal intensive care unit
- Author
-
Seema Bhangar, Brian Firek, Xiaochen Tang, Jillian F. Banfield, Brandon Brooks, Michael J. Morowitz, William W. Nazaroff, and Dusan Licina
- Subjects
Environmental Engineering ,Neonatal intensive care unit ,010504 meteorology & atmospheric sciences ,Particle number ,Geography, Planning and Development ,Environmental engineering ,Incubator ,Building and Construction ,010501 environmental sciences ,Particulates ,01 natural sciences ,Animal science ,Orders of magnitude (specific energy) ,Intensive care ,Particle-size distribution ,Particle ,Environmental science ,0105 earth and related environmental sciences ,Civil and Structural Engineering - Abstract
Infants in neonatal intensive care units (NICUs) are vulnerable to environmental stressors. Few studies have reported on airborne particles in the NICU environment. During a four-day pilot study in a private-style NICU, we measured size-resolved particle number (PN) concentrations with 1-min resolution. The investigation included simultaneous sampling in an unoccupied baby room and in an incubator of an otherwise normally functioning NICU. Background submicron (0.3–1 μm) particle levels in the room were 3–4 orders of magnitude lower than outdoors, owing to high-efficiency particulate filtration of supply air. Airborne supermicron particles were detected in the room; their presence was attributed primarily to emissions from occupant movements. The fraction of in-room PN detected within an infant incubator ranged from 0.2 for particles >10 μm to 0.6 for particles with diameter 0.3–0.5 μm. The incubator humidifier was a strong additional source of particles smaller than 5 μm. Activities by researchers, designed to simulate caregiver visits, were associated with elevated particle concentrations across all measured size ranges, and were particularly discernible among larger particles. Concentrations increased with the number of occupants and with the duration and vigor of activities. The highest levels were observed when fabrics were handled. Against the low background in this environment, even small occupancy-associated perturbations – such as from a brief entry – were discernible. Measurements from a second NICU in a different US region were found to be broadly similar. A notable difference was higher submicron particle levels in the second NICU, attributed to elevated outdoor pollution.
- Published
- 2016
- Full Text
- View/download PDF
48. Evidence for persistent and shared bacterial strains against a background of largely unique gut colonization in hospitalized premature infants
- Author
-
Tali Raveh-Sadka, Robyn Baker, Michael J. Morowitz, Christopher T. Brown, Brian C. Thomas, Itai Sharon, Jillian F. Banfield, and Brian Firek
- Subjects
Male ,0301 basic medicine ,Technology ,medicine.drug_class ,Antibiotics ,Low Birth Weight and Health of the Newborn ,2.2 Factors relating to physical environment ,Microbiology ,Enterococcus faecalis ,law.invention ,Cohort Studies ,Feces ,03 medical and health sciences ,Microbial ecology ,Clinical Research ,Preterm ,law ,Staphylococcus epidermidis ,Infant Mortality ,medicine ,Humans ,Colonization ,Sibling ,Premature ,Ecology, Evolution, Behavior and Systematics ,Pediatric ,Bacteria ,biology ,Infant, Newborn ,Infant ,Perinatal Period - Conditions Originating in Perinatal Period ,Biological Sciences ,Newborn ,biology.organism_classification ,Intensive care unit ,Gastrointestinal Microbiome ,Infectious Diseases ,030104 developmental biology ,Original Article ,Female ,Infection ,Infant, Premature ,Environmental Sciences - Abstract
© 2016 International Society for Microbial Ecology. All rights reserved. The potentially critical stage of initial gut colonization in premature infants occurs in the hospital environment, where infants are exposed to a variety of hospital-associated bacteria. Because few studies of microbial communities are strain-resolved, we know little about the extent to which specific strains persist in the hospital environment and disperse among infants. To study this, we compared 304 near-complete genomes reconstructed from fecal samples of 21 infants hospitalized in the same intensive care unit in two cohorts, over 3 years apart. The genomes represent 159 distinct bacterial strains, only 14 of which occurred in multiple infants. Enterococcus faecalis and Staphylococcus epidermidis, common infant gut colonists, exhibit diversity comparable to that of reference strains, inline with introduction of strains from infant-specific sources rather than a hospital strain pool. Unlike other infants, a pair of sibling infants shared multiple strains, even after extensive antibiotic administration, suggesting overlapping strain-sources and/or genetic selection drive microbiota similarities. Interestingly, however, five strains were detected in infants hospitalized three years apart. Three of these were also detected in multiple infants in the same year. This finding of a few widely dispersed and persistent bacterial colonizers despite overall low potential for strain dispersal among infants has implications for understanding and directing healthy colonization.
- Published
- 2016
- Full Text
- View/download PDF
49. Peroxisome Proliferator-activated Receptor-γ Coactivator 1-α (PGC1α) Protects against Experimental Murine Colitis
- Author
-
Garret Vincent, Kellie E. Cunningham, Michael J. Morowitz, Matthew B. Rogers, Chhinder P. Sodhi, Kevin P. Mollen, Donna B. Stolz, Sarangarajan Ranganathan, Brian S. Zuckerbraun, Elizabeth Novak, George K. Gittes, Charlotte E. Egan, David J. Hackam, and Brian Firek
- Subjects
0301 basic medicine ,Peroxisome proliferator-activated receptor ,Biology ,Biochemistry ,Inflammatory bowel disease ,Mice ,03 medical and health sciences ,Intestinal mucosa ,Coactivator ,medicine ,Animals ,Intestinal Mucosa ,Colitis ,Molecular Biology ,Barrier function ,Mice, Knockout ,chemistry.chemical_classification ,Mice, Inbred BALB C ,Dextran Sulfate ,Molecular Bases of Disease ,Cell Biology ,medicine.disease ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Intestinal epithelium ,Mitochondria ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Mitochondrial biogenesis ,Bacterial Translocation ,Immunology ,Transcription Factors - Abstract
Peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) is the primary regulator of mitochondrial biogenesis and was recently found to be highly expressed within the intestinal epithelium. PGC1α is decreased in the intestinal epithelium of patients with inflammatory bowel disease, but its role in pathogenesis is uncertain. We now hypothesize that PGC1α protects against the development of colitis and helps to maintain the integrity of the intestinal barrier. We selectively deleted PGC1α from the intestinal epithelium of mice by breeding a PGC1α(loxP/loxP) mouse with a villin-cre mouse. Their progeny (PGC1α(ΔIEC) mice) were subjected to 2% dextran sodium sulfate (DSS) colitis for 7 days. The SIRT1 agonist SRT1720 was used to enhance PGC1α activation in wild-type mice during DSS exposure. Mice lacking PGC1α within the intestinal epithelium were more susceptible to DSS colitis than their wild-type littermates. Pharmacologic activation of PGC1α successfully ameliorated disease and restored mitochondrial integrity. These findings suggest that a depletion of PGC1α in the intestinal epithelium contributes to inflammatory changes through a failure of mitochondrial structure and function as well as a breakdown of the intestinal barrier, which leads to increased bacterial translocation. PGC1α induction helps to maintain mitochondrial integrity, enhance intestinal barrier function, and decrease inflammation.
- Published
- 2016
- Full Text
- View/download PDF
50. Strategies to Promote ResiliencY (SPRY): a randomised embedded multifactorial adaptative platform (REMAP) clinical trial protocol to study interventions to improve recovery after surgery in high-risk patients
- Author
-
Derek C. Angus, Timothy D. Girard, Stephen A. Esper, Oscar C. Marroquin, Christopher W. Seymour, Scott M. Berry, Katherine M. Reitz, Matthew D. Neal, Alison Morris, Jason Kennedy, Brian S. Zuckerbraun, Anne B. Newman, Jennifer Vates, Barbara A. Methé, Kert Viele, Michelle A. Detry, Jennifer Holder-Murray, Michael J. Morowitz, and Melanie Quintana
- Subjects
medicine.medical_specialty ,Health Personnel ,Population ,Psychological intervention ,Postoperative Complications ,Interim ,Health care ,Humans ,Medicine ,adult surgery ,education ,Randomized Controlled Trials as Topic ,Protocol (science) ,clinical trials ,education.field_of_study ,business.industry ,Bayes Theorem ,information management ,General Medicine ,Perioperative ,Institutional review board ,Metformin ,Surgery ,Clinical trial ,business - Abstract
IntroductionAs the population ages, there is interest in strategies to promote resiliency, especially for frail patients at risk of its complications. The physiological stress of surgery in high-risk individuals has been proposed both as an important cause of accelerated age-related decline in health and as a model testing the effectiveness of strategies to improve resiliency to age-related health decline. We describe a randomised, embedded, multifactorial, adaptative platform (REMAP) trial to investigate multiple perioperative interventions, the first of which is metformin and selected for its anti-inflammatory and anti-ageing properties beyond its traditional blood glucose control features.Methods and analysisWithin a multihospital, single healthcare system, the Core Protocol for Strategies to Promote ResiliencY (SPRY) will be embedded within both the electronic health record (EHR) and the healthcare culture generating a continuously self-learning healthcare system. Embedding reduces the administrative burden of a traditional trial while accessing and rapidly analysing routine patient care EHR data. SPRY-Metformin is a placebo-controlled trial and is the first SPRY domain evaluating the effectiveness of three metformin dosages across three preoperative durations within a heterogeneous set of major surgical procedures. The primary outcome is 90-day hospital-free days. Bayesian posterior probabilities guide interim decision-making with predefined rules to determine stopping for futility or superior dosing selection. Using response adaptative randomisation, a maximum of 2500 patients allows 77%–92% power, detecting >15% primary outcome improvement. Secondary outcomes include mortality, readmission and postoperative complications. A subset of patients will be selected for substudies evaluating the microbiome, cognition, postoperative delirium and strength.Ethics and disseminationThe Core Protocol of SPRY REMAP and associated SPRY-Metformin Domain-Specific Appendix have been ethically approved by the Institutional Review Board and are publicly registered. Results will be publicly available to healthcare providers, patients and trial participants following achieving predetermined platform conclusions.Trial registration numberNCT03861767.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.