85 results on '"Manges AR"'
Search Results
2. Widespread distribution of urinary tract infections caused by a multidrug-resistant Escherichia coli clonal group.
- Author
-
Manges AR, Johnson JR, Foxman B, O'Bryan TT, Fullerton KE, Riley LW, Manges, A R, Johnson, J R, Foxman, B, O'Bryan, T T, Fullerton, K E, and Riley, L W
- Abstract
Background: The management of urinary tract infections is complicated by the increasing prevalence of antibiotic-resistant strains of Escherichia coli. We studied the clonal composition of E. coli isolates that were resistant to trimethoprim-sulfamethoxazole from women with community-acquired urinary tract infections.Methods: Prospectively collected E. coli isolates from women with urinary tract infections in a university community in California were evaluated for antibiotic susceptibility, O:H serotype, DNA fingerprinting, pulsed-field gel electrophoretic pattern, and virulence factors. The prevalence and characteristics of an antibiotic-resistant clone were evaluated in this group of isolates and in those from comparison cohorts in Michigan and Minnesota.Results: Fifty-five of the 255 E. coli isolates (22 percent) from the California cohort were resistant to trimethoprim-sulfamethoxazole as well as other antibiotics. There was a common pattern of DNA fingerprinting, suggesting that the isolates belonged to the same clonal group (clonal group A), in 28 of 55 isolates with trimethoprim-sulfamethoxazole resistance (51 percent) and in 2 of 50 randomly selected isolates that were susceptible to trimethoprim-sulfamethoxazole (4 percent, P<0.001). In addition, 11 of 29 resistant isolates (38 percent) from the Michigan cohort and 7 of 18 (39 percent) from the Minnesota cohort belonged to clonal group A. Most of the clonal group A isolates were serotype O11:H(nt) or O77:H(nt), with similar patterns of virulence factors, antibiotic susceptibility, and electrophoretic features.Conclusions: In three geographically diverse communities, a single clonal group accounted for nearly half of community-acquired urinary tract infections in women that were caused by E. coli strains with resistance to trimethoprim-sulfamethoxazole. The widespread distribution and high prevalence of E. coli clonal group A has major public health implications. [ABSTRACT FROM AUTHOR]- Published
- 2001
3. The effectiveness of colonoscopy in reducing mortality from colorectal cancer.
- Author
-
Romagnuolo J, Barkun AN, Manges AR, Romagnuolo, Joseph, Barkun, Alan N, and Manges, Amee R
- Published
- 2009
- Full Text
- View/download PDF
4. Extraintestinal E coli infections: reservoirs and transmission routes.
- Author
-
Manges AR
- Abstract
Extraintestinal infections with Escherichia coli cause significant morbidity and exact a high toll in health care costs. Urinary tract infection is the most common type of extraintestinal E coli infection, but bloodstream infections also occur. Environmental reservoirs of E coli have been implicated in the transmission of E coli causing human infections. Proposed transmission routes include person-to-person transmission, such as between sex partners and within households; transmission from companion animals to persons in the same household; and foodborne routes. Recent evidence suggests that food animal products, such as retail poultry, may serve as reservoirs of antimicrobial-resistant extraintestinal E coli. Evidence supports the sharing of E coli strains within households and among sex partners, but the transmission direction and dynamics have not yet been defined. [ABSTRACT FROM AUTHOR]
- Published
- 2007
5. Urinary tract infections and a multidrug-resistant Escherichia coli clonal group.
- Author
-
Sandel DC, Wang C, Kessler S, Petrof EO, Schwartz DN, Quinn JP, Barlam T, Moellering R, Johnson JR, Manges AR, and Riley LW
- Published
- 2002
6. Bifidobacterium longum and microbiome maturation modify a nutrient intervention for stunting in Zimbabwean infants.
- Author
-
Gough EK, Edens TJ, Carr L, Robertson RC, Mutasa K, Ntozini R, Chasekwa B, Geum HM, Baharmand I, Gill SK, Mutasa B, Mbuya MNN, Majo FD, Tavengwa N, Francis F, Tome J, Evans C, Kosek M, Prendergast AJ, and Manges AR
- Subjects
- Humans, Infant, Female, Male, Zimbabwe, Fucosyltransferases genetics, Feces microbiology, Bifidobacterium, Dietary Supplements, Nutrients, Gastrointestinal Microbiome, Growth Disorders prevention & control, Growth Disorders microbiology
- Abstract
Background: Small-quantity lipid-based nutrient supplements (SQ-LNS), which has been widely tested to reduce child stunting, has largely modest effects to date, but the mechanisms underlying these modest effects are unclear. Child stunting is a longstanding indicator of chronic undernutrition and it remains a prevalent public health problem. The infant gut microbiome may be a key contributor to stunting; and mother and infant fucosyltransferase (FUT) phenotypes are important determinants of infant microbiome composition., Methods: We investigated whether mother-infant FUT status (n = 792) and infant gut microbiome composition (n = 354 fecal specimens from 172 infants) modified the impact of an infant and young child feeding (IYCF) intervention, that included SQ-LNS, on stunting at age 18 months in secondary analysis of a randomized trial in rural Zimbabwe., Findings: We found that the impact of the IYCF intervention on stunting was modified by: (i) mother-infant FUT2+/FUT3- phenotype (difference-in-differences -32.6% [95% CI: -55.3%, -9.9%]); (ii) changes in species composition that reflected microbiome maturation (difference-in-differences -68.1% [95% CI: -99.0%, -28.5%); and (iii) greater relative abundance of B. longum (differences-in-differences 49.1% [95% CI: 26.6%, 73.6%]). The dominant strains of B. longum when the intervention started were most similar to the proficient milk oligosaccharide utilizer subspecies infantis, which decreased with infant age and differed by mother-infant FUT2+/FUT3- phenotypes., Interpretation: These findings indicate that a persistently "younger" microbiome at initiation of the intervention reduced its benefits on stunting in areas with a high prevalence of growth restriction., Funding: Bill and Melinda Gates Foundation, UK DFID/Aid, Wellcome Trust, Swiss Agency for Development and Cooperation, US National Institutes of Health, UNICEF, and Nutricia Research Foundation., Competing Interests: Declaration of interests AJP was supported by Wellcome Trust grant 108065/Z/15/Z. ARM was supported by Bill & Melinda Gates Foundation grant OPP1021542 and OPP1143707, with a subcontract to the University of British Columbia 20R25498 EKG was supported by The Nutricia Research Foundation grant 2021-52. T.J.E. was paid a scientific consulting fee in relation to the analysis of the data presented here by the Zvitambo Institute for Maternal and Child Health Research. RCR declares remittance from Abbott Nutrition Health Institute and Nutricia for public conference talks outside of the submitted work in the past 36 months. All other authors declare that they have no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. SinoNasal Microbiota Transfer to treat recalcitrant chronic rhinosinusitis: A case series.
- Author
-
Gill SK, Hernaiz-Leonardo JC, Edens TJ, Pascual A, Tang C, Fan J, Thamboo A, Mullings W, Alsaleh S, Alim BM, Javer AR, and Manges AR
- Subjects
- Humans, Chronic Disease, Middle Aged, Male, Female, Adult, Paranasal Sinuses microbiology, Treatment Outcome, Aged, Rhinosinusitis, Sinusitis microbiology, Sinusitis therapy, Rhinitis microbiology, Rhinitis therapy, Microbiota
- Abstract
Key Points: SinoNasal Microbiota Transfer (SNMT) was safe with immediate benefit in all recipients, with sustained improvement in two of three recipients for up to 180 days. The addition of antimicrobial photodynamic therapy worsened chronic rhinosinusitis. These promising SNMT results warrant further study of safety and efficacy., (© 2024 The Authors. International Forum of Allergy & Rhinology published by Wiley Periodicals LLC on behalf of American Academy of Otolaryngic Allergy and American Rhinologic Society.)
- Published
- 2024
- Full Text
- View/download PDF
8. Validation of an automated, end-to-end metagenomic sequencing assay for agnostic detection of respiratory viruses.
- Author
-
Gauthier NPG, Chan W, Locher K, Smailus D, Coope R, Charles M, Jassem A, Kopetzky J, Chorlton SD, and Manges AR
- Abstract
Background: Current molecular diagnostics are limited in the number and type of detectable pathogens. Metagenomic next generation sequencing (mNGS) is an emerging, and increasingly feasible, pathogen-agnostic diagnostic approach. Translational barriers prohibit the widespread adoption of this technology in clinical laboratories. We validate an end-to-end mNGS assay for detection of respiratory viruses. Our assay is optimized to reduce turnaround time, lower cost-per-sample, increase throughput, and deploy secure and actionable bioinformatic results., Methods: We validated our assay using residual nasopharyngeal swab specimens from Vancouver General Hospital (n = 359), RT-PCR-positive, or negative for Influenza, SARS-CoV-2, and RSV. We quantified sample stability, assay precision, the effect of background nucleic acid levels, and analytical limits of detection. Diagnostic performance metrics were estimated., Results: We report that our mNGS assay is highly precise, semi-quantitative, with analytical limits of detection ranging from 103-104 copies/mL. Our assay is highly specific (100%) and sensitive (61.9% Overall: 86.8%; RT-PCR Ct < 30). Multiplexing capabilities enable processing of up to 55-specimens simultaneously on an Oxford Nanopore GridION device, with results reported within 12-hours., Conclusions: This study outlines the diagnostic performance and feasibility of mNGS for respiratory viral diagnostics, infection control, and public health surveillance. We addressed translational barriers to widespread mNGS adoption., (© The Author(s) 2024. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)
- Published
- 2024
- Full Text
- View/download PDF
9. Bifidobacterium longum modifies a nutritional intervention for stunting in Zimbabwean infants.
- Author
-
Gough EK, Edens TJ, Carr L, Robertson RC, Mutasa K, Ntozini R, Chasekwa B, Geum HM, Baharmand I, Gill SK, Mutasa B, Mbuya MNN, Majo FD, Tavengwa N, Francis F, Tome J, Evans C, Kosek M, Prendergast AJ, and Manges AR
- Abstract
Child stunting is an indicator of chronic undernutrition and reduced human capital. However, it remains a poorly understood public health problem. Small-quantity lipid-based nutrient supplements (SQ-LNS) have been widely tested to reduce stunting, but have modest effects. The infant intestinal microbiome may contribute to stunting, and is partly shaped by mother and infant histo-blood group antigens (HBGA). We investigated whether mother-infant fucosyltransferase status, which governs HBGA, and the infant gut microbiome modified the impact of SQ-LNS on stunting at age 18 months among Zimbabwean infants in the SHINE Trial ( NCT01824940 ). We found that mother-infant fucosyltransferase discordance and Bifidobacterium longum reduced SQ-LNS efficacy. Infant age-related microbiome shifts in B. longum subspecies dominance from infantis , a proficient human milk oligosaccharide utilizer, to suis or longum , proficient plant-polysaccharide utilizers, were partly influenced by discordance in mother-infant FUT2+/FUT3- phenotype, suggesting that a "younger" microbiome at initiation of SQ-LNS reduces its benefits on stunting.
- Published
- 2024
- Full Text
- View/download PDF
10. Deficient butyrate metabolism in the intestinal microbiome is a potential risk factor for recurrent kidney stone disease.
- Author
-
Choy WH, Adler A, Morgan-Lang C, Gough EK, Hallam SJ, Manges AR, Chew BH, Penniston K, Miller A, and Lange D
- Subjects
- Humans, Oxalates metabolism, Risk Factors, Bacteria genetics, Butyrates, Gastrointestinal Microbiome, Kidney Calculi microbiology
- Abstract
Intestinal microbiome dysbiosis is a known risk factor for recurrent kidney stone disease (KSD) with prior data suggesting a role for dysfunctional metabolic pathways other than those directly utilizing oxalate. To identify alternative mechanisms, the current study analyzed differences in the metabolic potential of intestinal microbiomes of patients (n = 17) and live-in controls (n = 17) and determined their relevance to increased risk for KSD using shotgun metagenomic sequencing. We found no differences in the abundance of genes associated with known oxalate degradation pathways, supporting the notion that dysfunction in other metabolic pathways plays a role in KSD. Further analysis showed decreased abundance of key enzymes involved in butyrate biosynthesis in patient intestinal microbiomes. Furthermore, de novo construction of microbial genomes showed that the majority of genes significantly enriched in non-stone formers are affiliated with Faecalibacterium prausnitzii, a major butyrate producer. Specifically pertaining to butyrate metabolism, the majority of abundant genes mapped back to F. prausnitzii, Alistipes spp., and Akkermansia muciniphila. No differences were observed in ascorbate or glyoxylate metabolic pathways. Collectively, these data suggest that impaired bacterial-associated butyrate metabolism may be an oxalate-independent mechanism that contributes to an increased risk for recurrent KSD. This indicates that the role of the intestinal microbiome in recurrent KSD is multi-factorial, which is representative of the highly intertwined metabolic nature of this complex environment. Future bacteria-based treatments must not be restricted to targeting only oxalate metabolism., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
11. Treatment of HCV with direct-acting antivirals on reducing mortality related to extrahepatic manifestations: a large population-based study in British Columbia, Canada.
- Author
-
Jeong D, Wong S, Karim ME, Manges AR, Makuza JD, Bartlett SR, Velásquez García HA, Luster D, Adu PA, Binka M, Yu A, Krajden M, and Janjua NZ
- Abstract
Background: HCV infection is associated with mortality due to extrahepatic manifestations (EHM). Sustained virologic response (SVR) following direct-acting antiviral (DAA) therapy has been linked to decreased all-cause and liver-related mortality. However, evidence regarding the impact of DAA on EHM-related deaths is lacking. This study aimed to assess the impact of DAA and SVR on EHM-related mortality., Methods: The British Columbia Hepatitis Testers Cohort comprises ∼1.7 million people tested for HCV between 1990 and 2015 and is linked with administrative health data. Among individuals diagnosed with HCV by 12/31/2020, those who received at least one DAA treatment were matched to those who never received treatment by the year of their first HCV RNA positive date. We compared three groups: treated & SVR, treated & no-SVR, and untreated; and generated EHM mortality rates and incidence curves. To account for differences in baseline characteristics, we used inverse probability of treatment weights (IPTW). IPTW-weighted multivariable cause-specific Cox regression models were adjusted for competing risk and confounders., Findings: Study population included 12,815 treated (12,287 SVR, 528 no-SVR) and 12,815 untreated individuals (median follow-up 3.4 years, IQR 2.9). The untreated group had the highest EHM mortality rate (30.9 per 1000 person-years [PY], 95% CI 29.2-32.8), followed by the treated & no-SVR group (21.2 per 1000 PY, 95% CI 14.9-30.1), while the treated & SVR group had the lowest EHM mortality rate (7.9 per 1000 PY, 95% CI 7.1-8.7). In the multivariable model, EHM mortality in the treated & SVR group was significantly decreased (adjusted cause-specific hazard ratio [acsHR] 0.20, 95% CI 0.18-0.23). The treated & SVR group had significant reductions in mortality related to each of the EHMs (78-84%)., Interpretation: Treatment of HCV with DAA was associated with significant reductions in EHM-related mortality. These findings emphasize the critical importance of timely diagnosis and treatment of HCV to prevent deaths associated with EHM, and have important implications for clinical practice and public health., Funding: This work was supported by the BC Centre for Disease Control and the Canadian Institutes of Health Research (CIHR) [Grant # NHC-348216, PJT-156066, and PHE-337680]. DJ has received Doctoral Research Award (#201910DF1-435705-64343) from the Canadian Institutes of Health Research (CIHR) and Doctoral fellowship from the Canadian Network on Hepatitis C (CanHepC). CanHepC is funded by a joint initiative of the Canadian Institutes of Health Research (CIHR) (NHC-142832) and the Public Health Agency of Canada (PHAC)., Competing Interests: MK has received grant/research support from Roche, Merck, Siemens, Boeringer Ingelheim and Hologic. SRB has consulted for Cepheid, Gilead, and Abbvie, but no personal payments accepted, and has received investigator-initiated grants from Gilead and Abbvie through her institution. NZJ participated in advisory boards and has spoken for AbbVie and Gilead, not related to current work. DJ, SW, MEK, ARM, JDM, HAVG, DL, PA, MB and AY have no conflicts of interest to declare., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
12. Maternal vaginal microbiome composition does not affect development of the infant gut microbiome in early life.
- Author
-
Dos Santos SJ, Pakzad Z, Albert AYK, Elwood CN, Grabowska K, Links MG, Hutcheon JA, Maan EJ, Manges AR, Dumonceaux TJ, Hodgson ZG, Lyons J, Mitchell-Foster SM, Gantt S, Joseph KS, Van Schalkwyk JE, Hill JE, and Money DM
- Subjects
- Infant, Newborn, Humans, Infant, Pregnancy, Female, Prospective Studies, Canada, Feces microbiology, Gastrointestinal Microbiome genetics, Microbiota
- Abstract
Birth mode has been implicated as a major factor influencing neonatal gut microbiome development, and it has been assumed that lack of exposure to the maternal vaginal microbiome is responsible for gut dysbiosis among caesarean-delivered infants. Consequently, practices to correct dysbiotic gut microbiomes, such as vaginal seeding, have arisen while the effect of the maternal vaginal microbiome on that of the infant gut remains unknown. We conducted a longitudinal, prospective cohort study of 621 Canadian pregnant women and their newborn infants and collected pre-delivery maternal vaginal swabs and infant stool samples at 10-days and 3-months of life. Using cpn 60-based amplicon sequencing, we defined vaginal and stool microbiome profiles and evaluated the effect of maternal vaginal microbiome composition and various clinical variables on the development of the infant stool microbiome. Infant stool microbiomes showed significant differences in composition by delivery mode at 10-days postpartum; however, this effect could not be explained by maternal vaginal microbiome composition and was vastly reduced by 3 months. Vaginal microbiome clusters were distributed across infant stool clusters in proportion to their frequency in the overall maternal population, indicating independence of the two communities. Intrapartum antibiotic administration was identified as a confounder of infant stool microbiome differences and was associated with lower abundances of Escherichia coli , Bacteroides vulgatus , Bifidobacterium longum and Parabacteroides distasonis . Our findings demonstrate that maternal vaginal microbiome composition at delivery does not affect infant stool microbiome composition and development, suggesting that practices to amend infant stool microbiome composition focus factors other than maternal vaginal microbes., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Dos Santos, Pakzad, Albert, Elwood, Grabowska, Links, Hutcheon, Maan, Manges, Dumonceaux, Hodgson, Lyons, Mitchell-Foster, Gantt, Joseph, Van Schalkwyk, Hill and Money.)
- Published
- 2023
- Full Text
- View/download PDF
13. Agnostic Sequencing for Detection of Viral Pathogens.
- Author
-
Gauthier NPG, Chorlton SD, Krajden M, and Manges AR
- Subjects
- High-Throughput Nucleotide Sequencing methods, Bacteria genetics, Metagenomics methods, Genome, Viral genetics, Viruses genetics
- Abstract
The advent of next-generation sequencing (NGS) technologies has expanded our ability to detect and analyze microbial genomes and has yielded novel molecular approaches for infectious disease diagnostics. While several targeted multiplex PCR and NGS-based assays have been widely used in public health settings in recent years, these targeted approaches are limited in that they still rely on a priori knowledge of a pathogen's genome, and an untargeted or unknown pathogen will not be detected. Recent public health crises have emphasized the need to prepare for a wide and rapid deployment of an agnostic diagnostic assay at the start of an outbreak to ensure an effective response to emerging viral pathogens. Metagenomic techniques can nonspecifically sequence all detectable nucleic acids in a sample and therefore do not rely on prior knowledge of a pathogen's genome. While this technology has been reviewed for bacterial diagnostics and adopted in research settings for the detection and characterization of viruses, viral metagenomics has yet to be widely deployed as a diagnostic tool in clinical laboratories. In this review, we highlight recent improvements to the performance of metagenomic viral sequencing, the current applications of metagenomic sequencing in clinical laboratories, as well as the challenges that impede the widespread adoption of this technology.
- Published
- 2023
- Full Text
- View/download PDF
14. The gut microbiome and early-life growth in a population with high prevalence of stunting.
- Author
-
Robertson RC, Edens TJ, Carr L, Mutasa K, Gough EK, Evans C, Geum HM, Baharmand I, Gill SK, Ntozini R, Smith LE, Chasekwa B, Majo FD, Tavengwa NV, Mutasa B, Francis F, Tome J, Stoltzfus RJ, Humphrey JH, Prendergast AJ, and Manges AR
- Subjects
- Infant, Child, Humans, Child, Preschool, Prevalence, Growth Disorders epidemiology, Water Supply, Gastrointestinal Microbiome genetics, HIV Infections
- Abstract
Stunting affects one-in-five children globally and is associated with greater infectious morbidity, mortality and neurodevelopmental deficits. Recent evidence suggests that the early-life gut microbiome affects child growth through immune, metabolic and endocrine pathways. Using whole metagenomic sequencing, we map the assembly of the gut microbiome in 335 children from rural Zimbabwe from 1-18 months of age who were enrolled in the Sanitation, Hygiene, Infant Nutrition Efficacy Trial (SHINE; NCT01824940), a randomized trial of improved water, sanitation and hygiene (WASH) and infant and young child feeding (IYCF). Here, we show that the early-life gut microbiome undergoes programmed assembly that is unresponsive to the randomized interventions intended to improve linear growth. However, maternal HIV infection is associated with over-diversification and over-maturity of the early-life gut microbiome in their uninfected children, in addition to reduced abundance of Bifidobacterium species. Using machine learning models (XGBoost), we show that taxonomic microbiome features are poorly predictive of child growth, however functional metagenomic features, particularly B-vitamin and nucleotide biosynthesis pathways, moderately predict both attained linear and ponderal growth and growth velocity. New approaches targeting the gut microbiome in early childhood may complement efforts to combat child undernutrition., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Antibiotic use and resistance in children with severe acute malnutrition and human immunodeficiency virus infection.
- Author
-
Francis F, Robertson RC, Bwakura-Dangarembizi M, Prendergast AJ, and Manges AR
- Subjects
- Humans, Child, Infant, Anti-Bacterial Agents therapeutic use, HIV Infections complications, HIV Infections drug therapy, Severe Acute Malnutrition complications, Severe Acute Malnutrition therapy
- Abstract
Severe acute malnutrition (SAM) and human immunodeficiency virus (HIV) infection underlie a major proportion of the childhood disease burden in low- and middle-income countries. These diseases commonly co-occur and lead to higher risk of other endemic infectious diseases, thereby compounding the risk of mortality and morbidity. The widespread use of antibiotics as treatment and prophylaxis in childhood SAM and HIV infections, respectively, has reduced mortality and morbidity but canlead to increasing antibiotic resistance. Development of antibiotic resistance could render future infections untreatable. This review summarises the endemic co-occurrence of undernutrition, particularly SAM, and HIV in children, and current treatment practices, specifically WHO-recommended antibiotic usage. The risks and benefits of antibiotic treatment, prophylaxis and resistance are reviewed in the context of patients with SAM and HIV and associated sub-populations. Finally, the review highlights possible research areas and populations where antibiotic resistance progression can be studied to best address concerns associated with the future impact of resistance. Current antibiotic usage is lifesaving in complicated SAM and HIV-infected populations; nevertheless, increasing baseline resistance and infection remain a significant concern. In conclusion, antibiotic usage currently addresses the immediate needs of children in SAM and HIV endemic regions; however, it is prudent to evaluate the impact of antibiotic use on resistance dynamics and long-term child health., Competing Interests: Declarations of Competing Interests No competing interests to declare., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Alterations in the nasopharyngeal microbiome associated with SARS-CoV-2 infection status and disease severity.
- Author
-
Gauthier NPG, Locher K, MacDonald C, Chorlton SD, Charles M, and Manges AR
- Subjects
- Humans, Nasopharynx, Pandemics prevention & control, RNA, Ribosomal, 16S genetics, SARS-CoV-2, Severity of Illness Index, COVID-19, Microbiota
- Abstract
Objectives: The COVID-19 pandemic and ensuing public health emergency has emphasized the need to study SARS-CoV-2 pathogenesis. The human microbiome has been shown to regulate the host immune system and may influence host susceptibility to viral infection, as well as disease severity. Several studies have assessed whether compositional alterations in the nasopharyngeal microbiota are associated with SARS-CoV-2 infection. However, the results of these studies were varied, and many did not account for disease severity. This study aims to examine whether compositional differences in the nasopharyngeal microbiota are associated with SARS-CoV-2 infection status and disease severity., Methods: We performed Nanopore full-length 16S rRNA sequencing on 194 nasopharyngeal swab specimens from hospitalized and community-dwelling SARS-CoV-2-infected and uninfected individuals. Sequence data analysis was performed using the BugSeq 16S analysis pipeline., Results: We found significant beta (PERMANOVA p < 0.05), but not alpha (Kruskal-Wallis p > 0.05) diversity differences in the nasopharyngeal microbiota among our study groups. We identified several differentially abundant taxa associated with SARS-CoV-2 infection status and disease severity using ALDEx2. Finally, we observed a trend towards higher abundance of Enterobacteriaceae in specimens from hospitalized SARS-CoV-2-infected patients., Conclusions: This study identified several alterations in the nasopharyngeal microbiome associated with SARS-CoV-2 infection status and disease severity. Understanding the role of the microbiome in infection susceptibility and severity may open new avenues of research for disease prevention and treatment., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: SDC holds equity in BugSeq Bioinformatics Inc. This does not alter our adherence to PLoS ONE’s policies on sharing data and materials.
- Published
- 2022
- Full Text
- View/download PDF
17. BugSplit enables genome-resolved metagenomics through highly accurate taxonomic binning of metagenomic assemblies.
- Author
-
Chandrakumar I, Gauthier NPG, Nelson C, Bonsall MB, Locher K, Charles M, MacDonald C, Krajden M, Manges AR, and Chorlton SD
- Subjects
- Bacteria classification, COVID-19 epidemiology, COVID-19 prevention & control, COVID-19 virology, Humans, Internet, Pandemics prevention & control, Reproducibility of Results, SARS-CoV-2 classification, SARS-CoV-2 genetics, SARS-CoV-2 physiology, Algorithms, Bacteria genetics, Computational Biology methods, Metagenome genetics, Metagenomics methods, Nanopore Sequencing methods
- Abstract
A large gap remains between sequencing a microbial community and characterizing all of the organisms inside of it. Here we develop a novel method to taxonomically bin metagenomic assemblies through alignment of contigs against a reference database. We show that this workflow, BugSplit, bins metagenome-assembled contigs to species with a 33% absolute improvement in F1-score when compared to alternative tools. We perform nanopore mNGS on patients with COVID-19, and using a reference database predating COVID-19, demonstrate that BugSplit's taxonomic binning enables sensitive and specific detection of a novel coronavirus not possible with other approaches. When applied to nanopore mNGS data from cases of Klebsiella pneumoniae and Neisseria gonorrhoeae infection, BugSplit's taxonomic binning accurately separates pathogen sequences from those of the host and microbiota, and unlocks the possibility of sequence typing, in silico serotyping, and antimicrobial resistance prediction of each organism within a sample. BugSplit is available at https://bugseq.com/academic ., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. Virulence Genotype and Phenotype of Multiple Antimicrobial-Resistant Escherichia coli Isolates from Broilers Assessed from a "One-Health" Perspective.
- Author
-
Rehman MA, Rempel H, Carrillo CD, Ziebell K, Allen K, Manges AR, Topp E, and Diarra MS
- Subjects
- Animals, Anti-Bacterial Agents pharmacology, Caco-2 Cells, Genotype, Humans, Phenotype, Virulence genetics, Chickens microbiology, Drug Resistance, Multiple, Bacterial, Escherichia coli drug effects, Escherichia coli genetics, Escherichia coli pathogenicity
- Abstract
Abstract: Extraintestinal pathogenic Escherichia coli (ExPEC) include several serotypes that have been associated with colibacillosis in poultry and with urinary tract infections (UTIs) and newborn meningitis in humans. In this study, 57 antimicrobial-resistant E. coli from apparently healthy broiler chickens were characterized for their health and safety risks. These isolates belonged to 12 serotypes, and isolates of the same serotype were clonal based on single nucleotide variant analysis. Most of the isolates harbored plasmids; IncC and IncFIA were frequently detected. The majority of the resistant isolates harbored plasmid-mediated resistance genes, including aph(3″)-Ib, aph(6)-Id, blaCMY-2, floR, sul1, sul2, tet(A), and tet(B), in agreement with their resistant phenotypes. The class 1 integron was detected in all E. coli serotypes except O124:H25 and O7:H6. Of the 57 broiler E. coli isolates, 27 were avian pathogenic, among which 18 were also uropathogenic E. coli and the remainder were other ExPEC. The two isolates of serotype O161:H4 (ST117) were genetically related to the control avian pathogenic strains and a clinical isolate associated with UTIs. A strain of serotype O159:H45 (ST101) also was closely related to a UTI isolate. The detected virulence factors included adhesins, invasins, siderophores, type III secretion systems, and toxins in combination with other virulence determinants. A broiler isolate of serotype O7:H18 (ST38) carried the ibeA gene encoding a protein involved in invasion of brain endothelium on a 102-kbp genetic island. This isolate moderately adhered and invaded Caco-2 cells and induced mortality (42.5%) in a day-old-chick infection model. The results of this study suggest that multiple antimicrobial-resistant E. coli isolates recovered from apparent healthy broilers can be pathogenic and act as reservoirs for antimicrobial resistance genes, highlighting the necessity of their assessment in a "One-Heath" context., (Published 2022 by the International Association for Food Protection. Copyright ©, Her Majesty the Queen in Right of Canada, as represented by the Minister of Agriculture and Agri-Food Canada.)
- Published
- 2022
- Full Text
- View/download PDF
19. Optimization of magnetic bead-based nucleic acid extraction for SARS-CoV-2 testing using readily available reagents.
- Author
-
Haile S, Nikiforuk AM, Pandoh PK, Twa DDW, Smailus DE, Nguyen J, Pleasance S, Wong A, Zhao Y, Eisler D, Moksa M, Cao Q, Wong M, Su E, Krzywinski M, Nelson J, Mungall AJ, Tsang F, Prentice LM, Jassem A, Manges AR, Jones SJM, Coope RJ, Prystajecky N, Marra MA, Krajden M, and Hirst M
- Subjects
- COVID-19 Testing, Humans, Indicators and Reagents, Magnetic Phenomena, Pandemics, RNA, Viral genetics, SARS-CoV-2, Sensitivity and Specificity, COVID-19, Nucleic Acids
- Abstract
The COVID-19 pandemic has highlighted the need for generic reagents and flexible systems in diagnostic testing. Magnetic bead-based nucleic acid extraction protocols using 96-well plates on open liquid handlers are readily amenable to meet this need. Here, one such approach is rigorously optimized to minimize cross-well contamination while maintaining sensitivity., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Nanopore metagenomic sequencing for detection and characterization of SARS-CoV-2 in clinical samples.
- Author
-
Gauthier NPG, Nelson C, Bonsall MB, Locher K, Charles M, MacDonald C, Krajden M, Chorlton SD, and Manges AR
- Subjects
- Humans, Sensitivity and Specificity, COVID-19 diagnosis, Metagenome, Nanopore Sequencing methods, Pandemics, SARS-CoV-2 isolation & purification
- Abstract
Objectives: The COVID-19 pandemic has underscored the need for rapid novel diagnostic strategies. Metagenomic Next-Generation Sequencing (mNGS) may allow for the detection of pathogens that can be missed in targeted assays. The goal of this study was to assess the performance of nanopore-based Sequence-Independent Single Primer Amplification (SISPA) for the detection and characterization of SARS-CoV-2., Methods: We performed mNGS on clinical samples and designed a diagnostic classifier that corrects for barcode crosstalk between specimens. Phylogenetic analysis was performed on genome assemblies., Results: Our assay yielded 100% specificity overall and 95.2% sensitivity for specimens with a RT-PCR cycle threshold value less than 30. We assembled 10 complete, and one near-complete genomes from 20 specimens that were classified as positive by mNGS. Phylogenetic analysis revealed that 10/11 specimens from British Columbia had a closest relative to another British Columbian specimen. We found 100% concordance between phylogenetic lineage assignment and Variant of Concern (VOC) PCR results. Our assay was able to distinguish between the Alpha and Gamma variants, which was not possible with the current standard VOC PCR being used in British Columbia., Conclusions: This study supports future work examining the broader feasibility of nanopore mNGS as a diagnostic strategy for the detection and characterization of viral pathogens., Competing Interests: SDC holds equity in BugSeq Bioinformatics Inc. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2021
- Full Text
- View/download PDF
21. Early Neonatal Meconium Does Not Have a Demonstrable Microbiota Determined through Use of Robust Negative Controls with cpn 60-Based Microbiome Profiling.
- Author
-
Dos Santos SJ, Pakzad Z, Elwood CN, Albert AYK, Gantt S, Manges AR, Dumonceaux TJ, Maan EJ, Hill JE, and Money DM
- Subjects
- Adult, Bacteria genetics, Bacteria isolation & purification, Bacterial Load, Biomass, DNA, Bacterial genetics, Electrophoresis, Gel, Pulsed-Field, Enterococcus faecalis genetics, Enterococcus faecalis isolation & purification, Escherichia coli genetics, Escherichia coli isolation & purification, Female, Humans, Infant, Newborn, Male, Pregnancy, Skin microbiology, Staphylococcus epidermidis genetics, Staphylococcus epidermidis isolation & purification, Bacteria classification, DNA, Bacterial isolation & purification, Feces microbiology, Meconium microbiology, Microbiota genetics
- Abstract
Detection of bacterial DNA within meconium is often cited as evidence supporting in utero colonization. However, many studies fail to adequately control for contamination. We aimed to define the microbial content of meconium under properly controlled conditions. DNA was extracted from 141 meconium samples and subjected to cpn 60-based microbiome profiling, with controls to assess contamination throughout. Total bacterial loads of neonatal meconium, infant stool, and controls were compared by 16S rRNA quantitative PCR (qPCR). Viable bacteria within meconium were cultured, and isolate clonality was assessed by pulsed-field gel electrophoresis (PFGE). Meconium samples did not differ significantly from controls with respect to read numbers or taxonomic composition. Twenty (14%) outliers with markedly higher read numbers were collected significantly later after birth and appeared more like transitional stool than meconium. Total bacterial loads were significantly higher in stool than in meconium, which did not differ from that of sequencing controls, and correlated well with read numbers. Cultured isolates were most frequently identified as Staphylococcus epidermidis, Enterococcus faecalis, or Escherichia coli, with PFGE indicating high intraspecies diversity. Our findings highlight the importance of robust controls in studies of low microbial biomass samples and argue against meaningful bacterial colonization in utero . Given that meconium microbiome profiles could not be distinguished from sequencing controls, and that viable bacteria within meconium appeared uncommon and largely consistent with postnatal skin colonization, there does not appear to be a meconium microbiota. IMPORTANCE Much like the recent placental microbiome controversy, studies of neonatal meconium reporting bacterial communities within the fetal and neonatal gut imply that microbial colonization begins prior to birth. However, recent work has shown that placental microbiomes almost exclusively represent contamination from lab reagents and the environment. Here, we demonstrate that prior studies of neonatal meconium are impacted by the same issue, showing that the microbial content of meconium does not differ from negative controls that have never contained any biological material. Our culture findings similarly supported this notion and largely comprised bacteria normally associated with healthy skin. Overall, our work adds to the growing body of evidence against the in utero colonization hypothesis.
- Published
- 2021
- Full Text
- View/download PDF
22. The fecal microbiome and rotavirus vaccine immunogenicity in rural Zimbabwean infants.
- Author
-
Robertson RC, Church JA, Edens TJ, Mutasa K, Min Geum H, Baharmand I, Gill SK, Ntozini R, Chasekwa B, Carr L, Majo FD, Kirkpatrick BD, Lee B, Moulton LH, Humphrey JH, Prendergast AJ, and Manges AR
- Subjects
- Antibodies, Viral, Humans, Immunogenicity, Vaccine, Immunoglobulin A, Infant, Gastrointestinal Microbiome, Rotavirus genetics, Rotavirus Infections prevention & control, Rotavirus Vaccines
- Abstract
Background: Oral rotavirus vaccine (RVV) immunogenicity is considerably lower in low- versus high-income populations; however, the mechanisms underlying this remain unclear. Previous evidence suggests that the gut microbiota may contribute to differences in oral vaccine efficacy., Methods: We performed whole metagenome shotgun sequencing on stool samples and measured anti-rotavirus immunoglobulin A in plasma samples from a subset of infants enrolled in a cluster randomized 2 × 2 factorial trial of improved water, sanitation and hygiene and infant feeding in rural Zimbabwe (SHINE trial: NCT01824940). We examined taxonomic microbiome composition and functional metagenome features using random forest models, differential abundance testing and regression analyses to explored associations with RVV immunogenicity., Results: Among 158 infants with stool samples and anti-rotavirus IgA titres, 34 were RVV seroconverters. The median age at stool collection was 43 days (IQR: 35-68), corresponding to a median of 4 days before the first RVV dose. The infant microbiome was dominated by Bifidobacterium longum. The gut microbiome differed significantly between early (≤42 days) and later samples (>42 days) however, we observed no meaningful differences in alpha diversity, beta diversity, species composition or functional metagenomic features by RVV seroconversion status. Bacteroides thetaiotaomicron was the only species associated with anti-rotavirus IgA titre. Random forest models poorly classified seroconversion status by both composition and functional microbiome variables., Conclusions: RVV immunogenicity is low in this rural Zimbabwean setting, however it was not associated with the composition or function of the early-life gut microbiome in this study. Further research is warranted to examine the mechanisms of poor oral RVV efficacy in low-income countries., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Maternal fecal microbiome predicts gestational age, birth weight and neonatal growth in rural Zimbabwe.
- Author
-
Gough EK, Edens TJ, Geum HM, Baharmand I, Gill SK, Robertson RC, Mutasa K, Ntozini R, Smith LE, Chasekwa B, Majo FD, Tavengwa NV, Mutasa B, Francis F, Carr L, Tome J, Stoltzfus RJ, Moulton LH, Prendergast AJ, Humphrey JH, Manges AR, and Team ST
- Subjects
- Bacteria genetics, Bacteria isolation & purification, Child Development, Female, Gestational Age, High-Throughput Nucleotide Sequencing, Humans, Infant, Newborn, Male, Mothers, Pregnancy, Randomized Controlled Trials as Topic, Rural Population, Sequence Analysis, DNA, Zimbabwe, Bacteria classification, Birth Weight, Body Height, Feces microbiology, Metagenomics methods, RNA, Ribosomal, 16S genetics
- Abstract
Background: Preterm birth and low birth weight (LBW) affect one in ten and one in seven livebirths, respectively, primarily in low-income and middle-income countries (LMIC) and are major predictors of poor child health outcomes. However, both have been recalcitrant to public health intervention. The maternal intestinal microbiome may undergo substantial changes during pregnancy and may influence fetal and neonatal health in LMIC populations., Methods: Within a subgroup of 207 mothers and infants enrolled in the SHINE trial in rural Zimbabwe, we performed shotgun metagenomics on 351 fecal specimens provided during pregnancy and at 1-month post-partum to investigate the relationship between the pregnancy gut microbiome and infant gestational age, birth weight, 1-month length-, and weight-for-age z-scores using extreme gradient boosting machines., Findings: Pregnancy gut microbiome taxa and metabolic functions predicted birth weight and WAZ at 1 month more accurately than gestational age and LAZ. Blastoscystis sp, Brachyspira sp and Treponeme carriage were high compared to Western populations. Resistant starch-degraders were important predictors of birth outcomes. Microbiome capacity for environmental sensing, vitamin B metabolism, and signalling predicted increased infant birth weight and neonatal growth; while functions involved in biofilm formation in response to nutrient starvation predicted reduced birth weight and growth., Interpretation: The pregnancy gut microbiome in rural Zimbabwe is characterized by resistant starch-degraders and may be an important metabolic target to improve birth weight., Funding: Bill and Melinda Gates Foundation, UK Department for International Development, Wellcome Trust, Swiss Agency for Development and Cooperation, US National Institutes of Health, and UNICEF., Competing Interests: Declaration of Competing Interest None., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Can breastfeeding protect against antimicrobial resistance?
- Author
-
Nadimpalli ML, Bourke CD, Robertson RC, Delarocque-Astagneau E, Manges AR, and Pickering AJ
- Subjects
- Female, Humans, Infant, Newborn, Male, Breast Feeding methods, Drug Resistance, Bacterial physiology, Gastrointestinal Microbiome immunology, Milk, Human microbiology
- Abstract
Background: The proportion of infections among young children that are antimicrobial-resistant is increasing across the globe. Newborns may be colonized with enteric antimicrobial-resistant pathogens early in life, which is a risk factor for infection-related morbidity and mortality. Breastfeeding is actively promoted worldwide for its beneficial impacts on newborn health and gut health. However, the role of breastfeeding and human milk components in mitigating young children's carriage of antimicrobial-resistant pathogens and antibiotic resistance genes has not been comprehensively explored., Main Body: Here, we review how the act of breastfeeding, early breastfeeding, and/or human milk components, such as the milk microbiota, secretory IgA, human milk oligosaccharides, antimicrobial peptides, and microRNA -bearing extracellular vesicles, could play a role in preventing the establishment of antimicrobial-resistant pathogens in young children's developing gut microbiomes. We describe findings from recent human studies that support this concept., Conclusion: Given the projected rise in global morbidity and mortality that will stem from antimicrobial-resistant infections, identifying behavioral or nutritional interventions that could decrease children's susceptibility to colonization with antimicrobial-resistant pathogens may be one strategy for protecting their health. We suggest that breastfeeding and human milk supplements deserve greater attention as potential preventive measures in the global effort to combat antimicrobial resistance, particularly in low- and middle-income settings.
- Published
- 2020
- Full Text
- View/download PDF
25. Brief Report: Cessation of Long-Term Cotrimoxazole Prophylaxis in HIV-Infected Children Does Not Alter the Carriage of Antimicrobial Resistance Genes.
- Author
-
Francis F, Gough EK, Edens TJ, Berejena C, Bwakura-Dangarembizi M, Shonhai A, Nathoo KJ, Glass M, Gibb DM, Prendergast AJ, and Manges AR
- Subjects
- Anti-Bacterial Agents pharmacology, Anti-Infective Agents administration & dosage, Child, Child, Preschool, Drug Resistance, Microbial genetics, Female, Gastrointestinal Microbiome genetics, Humans, Male, Trimethoprim, Sulfamethoxazole Drug Combination administration & dosage, Zimbabwe, Anti-Infective Agents therapeutic use, Drug Resistance, Microbial drug effects, Gastrointestinal Microbiome drug effects, HIV Infections complications, Trimethoprim, Sulfamethoxazole Drug Combination therapeutic use
- Abstract
Background: Cotrimoxazole (CTX) is a broad-spectrum antimicrobial, combining trimethoprim and sulfamethoxazole. CTX prophylaxis reduces mortality and morbidity among people living with HIV in regions with high prevalence of bacterial infections and malaria. The Antiretroviral research for Watoto trial evaluated the effect of stopping versus continuing CTX prophylaxis in sub-Saharan Africa., Methods: In this study, 72 HIV-infected Zimbabwean children, on antiretroviral therapy, provided fecal samples at 84 and 96 weeks after randomization to continue or stop CTX. DNA was extracted for whole metagenome shotgun sequencing, with sequencing reads mapped to the Comprehensive Antibiotic Resistance Database to identify CTX and other antimicrobial resistance genes., Results: There were minimal differences in the carriage of CTX resistance genes between groups. The dfrA1 gene, conferring trimethoprim resistance, was significantly higher in the continue group (P = 0.039) and the tetA(P) gene conferring resistance to tetracycline was significantly higher in the stop group (P = 0.013). CTX prophylaxis has a role in shaping the resistome; however, stopping prophylaxis does not decrease resistance gene abundance., Conclusions: No differences were observed in resistance gene carriage between the stop and continue groups. The previously shown multi-faceted protective effects of CTX in antiretroviral research for Watoto trial clinical outcomes are not outweighed by the risk of multi-drug resistance gene selection due to prophylaxis. These findings are reassuring, given current recommendations for long-term CTX prophylaxis among children living with HIV in sub-Saharan Africa to decrease mortality and morbidity.
- Published
- 2020
- Full Text
- View/download PDF
26. Strain-level analysis of gut-resident pro-inflammatory viridans group Streptococci suppressed by long-term cotrimoxazole prophylaxis among HIV-positive children in Zimbabwe.
- Author
-
Gough EK, Bourke CD, Berejena C, Shonhai A, Bwakura-Dangarembizi M, Prendergast AJ, and Manges AR
- Subjects
- Anti-Bacterial Agents therapeutic use, Anti-HIV Agents therapeutic use, Child, Feces microbiology, HIV Infections drug therapy, Humans, Inflammation microbiology, Inflammation prevention & control, Intestines immunology, Intestines microbiology, Species Specificity, Streptococcus salivarius classification, Streptococcus salivarius physiology, Viridans Streptococci classification, Viridans Streptococci physiology, Zimbabwe, Antibiotic Prophylaxis, Gastrointestinal Microbiome drug effects, HIV Infections microbiology, Streptococcus salivarius drug effects, Trimethoprim, Sulfamethoxazole Drug Combination therapeutic use, Viridans Streptococci drug effects
- Abstract
Antimicrobials have become a mainstay of healthcare in the past century due to their activity against pathogens. More recently, it has become clear that they can also affect health via their impact on the microbiota and inflammation. This may explain some of their clinical benefits despite global increases in antimicrobial resistance (AMR) and reduced antimicrobial effectiveness. We showed in a randomized controlled trial of stopping versus continuing cotrimoxazole prophylaxis among HIV-positive Zimbabwean children taking antiretroviral therapy (ART), that continuation of cotrimoxazole persistently suppressed gut-resident viridans group streptococcal species (VGS) that were associated with intestinal inflammation. In this addendum, we provide a broader overview of how antibiotics can shape the microbiota and use high read-depth whole metagenome sequencing data from our published study to investigate whether (i) the impact of cotrimoxazole on gut VGS and (ii) VGS associated inflammation, is attributable to strain-level variability. We focus on S. salivarius , the VGS species that was most prevalent in the cohort and for which there was sufficient genome coverage to differentiate strains. We demonstrate that suppression of S. salivarius by cotrimoxazole is not strain specific, nor did stool concentration of the pro-inflammatory mediator myeloperoxidase vary by S. salivarius strain. We also show that gut-resident S. salivarius strains present in this study population are distinct from common oral strains. This is the first analysis of how cotrimoxazole prophylaxis used according to international treatment guidelines for children living with HIV influences the gut microbiome at the strain-level. We also provide a detailed review of the literature on the mechanisms by which suppression of VGS may act synergistically with cotrimoxazole's anti-inflammatory effects to reduce gut inflammation. A greater understanding of the sub-clinical effects of antibiotics offers new insights into their responsible clinical use.
- Published
- 2020
- Full Text
- View/download PDF
27. Escherichia coli causing bloodstream and other extraintestinal infections: tracking the next pandemic.
- Author
-
Manges AR
- Subjects
- England, Humans, Pandemics, Scotland, Wales, beta-Lactamases, Escherichia coli, Escherichia coli Infections epidemiology
- Published
- 2019
- Full Text
- View/download PDF
28. Genomic Epidemiology of Major Extraintestinal Pathogenic Escherichia coli Lineages Causing Urinary Tract Infections in Young Women Across Canada.
- Author
-
Fibke CD, Croxen MA, Geum HM, Glass M, Wong E, Avery BP, Daignault D, Mulvey MR, Reid-Smith RJ, Parmley EJ, Portt A, Boerlin P, and Manges AR
- Abstract
Background: A few extraintestinal pathogenic Escherichia coli (ExPEC) multilocus sequence types (STs) cause the majority of community-acquired urinary tract infections (UTIs). We examine the genomic epidemiology of major ExPEC lineages, specifically factors associated with intestinal acquisition., Methods: A total of 385 women with UTI caused by E. coli across Canada were asked about their diet, travel, and other exposures. Genome sequencing was used to determine both ST and genomic similarity. Logistic regression was used to identify factors associated with the acquisition of and infection with major ExPEC STs relative to minor ExPEC STs., Results: ST131, ST69, ST73, ST127, and ST95 were responsible for 54% of all UTIs. Seven UTI clusters were identified, but genomes from the ST95, ST127, and ST420 clusters exhibited as few as 3 single nucleotide variations across the entire genome, suggesting recent acquisition. Furthermore, we identified a cluster of UTIs caused by 6 genetically-related ST1193 isolates carrying mutations in gyrA and parC . The acquisition of and infection with ST69, ST95, ST127, and ST131 were all associated with increased travel. The consumption of high-risk foods such as raw meat or vegetables, undercooked eggs, and seafood was associated with acquisition of and infection with ST69, ST127, and ST131, respectively., Conclusions: Reservoirs may aid in the dissemination of pandemic ExPEC lineages in the community. Identifying ExPEC reservoirs may help prevent future emergence and dissemination of high-risk lineages within the community setting., (© The Author(s) 2019. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)
- Published
- 2019
- Full Text
- View/download PDF
29. Global Extraintestinal Pathogenic Escherichia coli (ExPEC) Lineages.
- Author
-
Manges AR, Geum HM, Guo A, Edens TJ, Fibke CD, and Pitout JDD
- Subjects
- Drug Resistance, Bacterial, Escherichia coli Infections epidemiology, Escherichia coli Infections prevention & control, Extraintestinal Pathogenic Escherichia coli classification, Extraintestinal Pathogenic Escherichia coli pathogenicity, Genome, Bacterial genetics, Humans, Escherichia coli Infections microbiology, Extraintestinal Pathogenic Escherichia coli genetics
- Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) strains are responsible for a majority of human extraintestinal infections globally, resulting in enormous direct medical and social costs. ExPEC strains are comprised of many lineages, but only a subset is responsible for the vast majority of infections. Few systematic surveillance systems exist for ExPEC. To address this gap, we systematically reviewed and meta-analyzed 217 studies (1995 to 2018) that performed multilocus sequence typing or whole-genome sequencing to genotype E. coli recovered from extraintestinal infections or the gut. Twenty major ExPEC sequence types (STs) accounted for 85% of E. coli isolates from the included studies. ST131 was the most common ST from 2000 onwards, covering all geographic regions. Antimicrobial resistance-based isolate study inclusion criteria likely led to an overestimation and underestimation of some lineages. European and North American studies showed similar distributions of ExPEC STs, but Asian and African studies diverged. Epidemiology and population dynamics of ExPEC are complex; summary proportion for some STs varied over time (e.g., ST95), while other STs were constant (e.g., ST10). Persistence, adaptation, and predominance in the intestinal reservoir may drive ExPEC success. Systematic, unbiased tracking of predominant ExPEC lineages will direct research toward better treatment and prevention strategies for extraintestinal infections., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
30. Cotrimoxazole reduces systemic inflammation in HIV infection by altering the gut microbiome and immune activation.
- Author
-
Bourke CD, Gough EK, Pimundu G, Shonhai A, Berejena C, Terry L, Baumard L, Choudhry N, Karmali Y, Bwakura-Dangarembizi M, Musiime V, Lutaakome J, Kekitiinwa A, Mutasa K, Szubert AJ, Spyer MJ, Deayton JR, Glass M, Geum HM, Pardieu C, Gibb DM, Klein N, Edens TJ, Walker AS, Manges AR, and Prendergast AJ
- Subjects
- CD4-Positive T-Lymphocytes immunology, Child, Child, Preschool, Cytokines metabolism, Disease Progression, Epithelial Cells drug effects, Epithelial Cells metabolism, HIV Infections immunology, Humans, Inflammation Mediators metabolism, Intestines drug effects, Intestines pathology, Nutritional Status drug effects, Phenotype, Streptococcus drug effects, Trimethoprim, Sulfamethoxazole Drug Combination pharmacology, Gastrointestinal Microbiome drug effects, HIV Infections drug therapy, HIV Infections microbiology, Inflammation drug therapy, Inflammation immunology, Trimethoprim, Sulfamethoxazole Drug Combination therapeutic use
- Abstract
Long-term cotrimoxazole prophylaxis reduces mortality and morbidity in HIV infection, but the mechanisms underlying these clinical benefits are unclear. Here, we investigate the impact of cotrimoxazole on systemic inflammation, an independent driver of HIV mortality. In HIV-positive Ugandan and Zimbabwean children receiving antiretroviral therapy, we show that plasma inflammatory markers were lower after randomization to continue ( n = 144) versus stop ( n = 149) cotrimoxazole. This was not explained by clinical illness, HIV progression, or nutritional status. Because subclinical enteropathogen carriage and enteropathy can drive systemic inflammation, we explored cotrimoxazole effects on the gut microbiome and intestinal inflammatory biomarkers. Although global microbiome composition was unchanged, viridans group Streptococci and streptococcal mevalonate pathway enzymes were lower among children continuing ( n = 36) versus stopping ( n = 36) cotrimoxazole. These changes were associated with lower fecal myeloperoxidase. To isolate direct effects of cotrimoxazole on immune activation from antibiotic effects, we established in vitro models of systemic and intestinal inflammation. In vitro cotrimoxazole had modest but consistent inhibitory effects on proinflammatory cytokine production by blood leukocytes from HIV-positive ( n = 16) and HIV-negative ( n = 8) UK adults and reduced IL-8 production by gut epithelial cell lines. Collectively we demonstrate that cotrimoxazole reduces systemic and intestinal inflammation both indirectly via antibiotic effects on the microbiome and directly by blunting immune and epithelial cell activation. Synergy between these pathways may explain the clinical benefits of cotrimoxazole despite high antimicrobial resistance, providing further rationale for extending coverage among people living with HIV in sub-Saharan Africa., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
31. The Human Microbiome and Child Growth - First 1000 Days and Beyond.
- Author
-
Robertson RC, Manges AR, Finlay BB, and Prendergast AJ
- Subjects
- Bacteria classification, Child, Preschool, Environment, Female, Gastrointestinal Microbiome immunology, Gastrointestinal Microbiome physiology, Growth and Development immunology, Humans, Infant, Infant, Newborn, Infections, Malnutrition, Pregnancy, Gastrointestinal Tract microbiology, Growth and Development physiology, Host Microbial Interactions immunology, Host Microbial Interactions physiology, Microbiota immunology, Microbiota physiology
- Abstract
The assembly of microbial communities within the gastrointestinal tract during early life plays a critical role in immune, endocrine, metabolic, and other host developmental pathways. Environmental insults during this period, such as food insecurity and infections, can disrupt this optimal microbial succession, which may contribute to lifelong and intergenerational deficits in growth and development. Here, we review the human microbiome in the first 1000 days - referring to the period from conception to 2 years of age - and using a developmental model, we examine the role of early microbial succession in growth and development. We propose that an 'undernourished' microbiome is intergenerational, thereby perpetuating growth impairments into successive generations. We also identify and discuss the intertwining host-microbe-environment interactions occurring prenatally and during early infancy, which may impair the trajectories of healthy growth and development, and explore their potential as novel microbial targets for intervention., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
32. Health Outcomes, Pathogenesis and Epidemiology of Severe Acute Malnutrition (HOPE-SAM): rationale and methods of a longitudinal observational study.
- Author
-
Bwakura-Dangarembizi M, Amadi B, Bourke CD, Robertson RC, Mwapenya B, Chandwe K, Kapoma C, Chifunda K, Majo F, Ngosa D, Chakara P, Chulu N, Masimba F, Mapurisa I, Besa E, Mutasa K, Mwakamui S, Runodamoto T, Humphrey JH, Ntozini R, Wells JCK, Manges AR, Swann JR, Walker AS, Nathoo KJ, Kelly P, and Prendergast AJ
- Subjects
- Child, Preschool, Female, HIV Infections complications, Humans, Infant, Infant, Newborn, Kaplan-Meier Estimate, Linear Models, Longitudinal Studies, Male, Prevalence, ROC Curve, Recurrence, Risk Factors, Zambia epidemiology, Zimbabwe epidemiology, Child Development, Patient Discharge statistics & numerical data, Severe Acute Malnutrition mortality, Severe Acute Malnutrition therapy
- Abstract
Introduction: Mortality among children hospitalised for complicated severe acute malnutrition (SAM) remains high despite the implementation of WHO guidelines, particularly in settings of high HIV prevalence. Children continue to be at high risk of morbidity, mortality and relapse after discharge from hospital although long-term outcomes are not well documented. Better understanding the pathogenesis of SAM and the factors associated with poor outcomes may inform new therapeutic interventions., Methods and Analysis: The Health Outcomes, Pathogenesis and Epidemiology of Severe Acute Malnutrition (HOPE-SAM) study is a longitudinal observational cohort that aims to evaluate the short-term and long-term clinical outcomes of HIV-positive and HIV-negative children with complicated SAM, and to identify the risk factors at admission and discharge from hospital that independently predict poor outcomes. Children aged 0-59 months hospitalised for SAM are being enrolled at three tertiary hospitals in Harare, Zimbabwe and Lusaka, Zambia. Longitudinal mortality, morbidity and nutritional data are being collected at admission, discharge and for 48 weeks post discharge. Nested laboratory substudies are exploring the role of enteropathy, gut microbiota, metabolomics and cellular immune function in the pathogenesis of SAM using stool, urine and blood collected from participants and from well-nourished controls., Ethics and Dissemination: The study is approved by the local and international institutional review boards in the participating countries (the Joint Research Ethics Committee of the University of Zimbabwe, Medical Research Council of Zimbabwe and University of Zambia Biomedical Research Ethics Committee) and the study sponsor (Queen Mary University of London). Caregivers provide written informed consent for each participant. Findings will be disseminated through peer-reviewed journals, conference presentations and to caregivers at face-to-face meetings., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2019
- Full Text
- View/download PDF
33. Independent and combined effects of improved water, sanitation, and hygiene, and improved complementary feeding, on child stunting and anaemia in rural Zimbabwe: a cluster-randomised trial.
- Author
-
Humphrey JH, Mbuya MNN, Ntozini R, Moulton LH, Stoltzfus RJ, Tavengwa NV, Mutasa K, Majo F, Mutasa B, Mangwadu G, Chasokela CM, Chigumira A, Chasekwa B, Smith LE, Tielsch JM, Jones AD, Manges AR, Maluccio JA, and Prendergast AJ
- Subjects
- Adult, Child, Preschool, Female, Humans, Infant, Male, Pregnancy, Zimbabwe epidemiology, Anemia epidemiology, Growth Disorders epidemiology, Hygiene standards, Infant Nutritional Physiological Phenomena standards, Rural Health statistics & numerical data, Sanitation standards, Water Supply standards
- Abstract
Background: Child stunting reduces survival and impairs neurodevelopment. We tested the independent and combined effects of improved water, sanitation, and hygiene (WASH), and improved infant and young child feeding (IYCF) on stunting and anaemia in in Zimbabwe., Methods: We did a cluster-randomised, community-based, 2 × 2 factorial trial in two rural districts in Zimbabwe. Clusters were defined as the catchment area of between one and four village health workers employed by the Zimbabwe Ministry of Health and Child Care. Women were eligible for inclusion if they permanently lived in clusters and were confirmed pregnant. Clusters were randomly assigned (1:1:1:1) to standard of care (52 clusters), IYCF (20 g of a small-quantity lipid-based nutrient supplement per day from age 6 to 18 months plus complementary feeding counselling; 53 clusters), WASH (construction of a ventilated improved pit latrine, provision of two handwashing stations, liquid soap, chlorine, and play space plus hygiene counselling; 53 clusters), or IYCF plus WASH (53 clusters). A constrained randomisation technique was used to achieve balance across the groups for 14 variables related to geography, demography, water access, and community-level sanitation coverage. Masking of participants and fieldworkers was not possible. The primary outcomes were infant length-for-age Z score and haemoglobin concentrations at 18 months of age among children born to mothers who were HIV negative during pregnancy. These outcomes were analysed in the intention-to-treat population. We estimated the effects of the interventions by comparing the two IYCF groups with the two non-IYCF groups and the two WASH groups with the two non-WASH groups, except for outcomes that had an important statistical interaction between the interventions. This trial is registered with ClinicalTrials.gov, number NCT01824940., Findings: Between Nov 22, 2012, and March 27, 2015, 5280 pregnant women were enrolled from 211 clusters. 3686 children born to HIV-negative mothers were assessed at age 18 months (884 in the standard of care group from 52 clusters, 893 in the IYCF group from 53 clusters, 918 in the WASH group from 53 clusters, and 991 in the IYCF plus WASH group from 51 clusters). In the IYCF intervention groups, the mean length-for-age Z score was 0·16 (95% CI 0·08-0·23) higher and the mean haemoglobin concentration was 2·03 g/L (1·28-2·79) higher than those in the non-IYCF intervention groups. The IYCF intervention reduced the number of stunted children from 620 (35%) of 1792 to 514 (27%) of 1879, and the number of children with anaemia from 245 (13·9%) of 1759 to 193 (10·5%) of 1845. The WASH intervention had no effect on either primary outcome. Neither intervention reduced the prevalence of diarrhoea at 12 or 18 months. No trial-related serious adverse events, and only three trial-related adverse events, were reported., Interpretation: Household-level elementary WASH interventions implemented in rural areas in low-income countries are unlikely to reduce stunting or anaemia and might not reduce diarrhoea. Implementation of these WASH interventions in combination with IYCF interventions is unlikely to reduce stunting or anaemia more than implementation of IYCF alone., Funding: Bill & Melinda Gates Foundation, UK Department for International Development, Wellcome Trust, Swiss Development Cooperation, UNICEF, and US National Institutes of Health., (Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
34. Self-reported risk factors for having Escherichia coli sequence type 131 or its H 30 subclone among US Veterans with a clinical E. coli isolate.
- Author
-
Manges AR, Thuras P, Porter S, and Johnson JR
- Abstract
Among 469 US military veterans with an Escherichia coli clinical isolate (2012-2013), we explored healthcare and non-healthcare risk factors for having E. coli sequence type 131 and its H30 subclone (ST131-H30). Overall, 66 (14%) isolates were ST131; 51 (77%) of these were ST131-H30. After adjustment for healthcare-associated factors, ST131 remained positively associated with medical lines and nursing home residence. After adjustment for environmental factors, ST131 remained associated with wild animal contact (positive), meat consumption (negative) and pet cat exposure (negative). Thus, ST131 was associated predominantly with healthcare-associated exposures, while non-ST131 E. coli were associated with some environmental exposures.
- Published
- 2018
- Full Text
- View/download PDF
35. Reply to Davido et al.
- Author
-
Leung V, Vincent C, Edens TJ, Miller MA, and Manges AR
- Subjects
- Anti-Bacterial Agents, Drug Resistance, Bacterial, Fecal Microbiota Transplantation, Humans, Anti-Infective Agents, Clostridium Infections
- Published
- 2018
- Full Text
- View/download PDF
36. Environmental enteric dysfunction pathways and child stunting: A systematic review.
- Author
-
Harper KM, Mutasa M, Prendergast AJ, Humphrey J, and Manges AR
- Subjects
- Child, Enteritis pathology, Humans, Bacterial Translocation, Biomarkers analysis, Enteritis complications, Growth Disorders
- Abstract
Background: Environmental enteric dysfunction (EED) is commonly defined as an acquired subclinical disorder of the small intestine, characterized by villous atrophy and crypt hyperplasia. EED has been proposed to underlie stunted growth among children in developing countries. A collection of biomarkers, organized into distinct domains, has been used to measure different aspects of EED. Here, we examine whether these hypothesized relationships, among EED domains and between each domain and stunting, are supported by data from recent studies., Methodology: A systematic literature search was conducted using PubMed, MEDLINE, EMBASE, Web of Science, and CINAHL between January 1, 2010 and April 20, 2017. Information on study objective, design, population, location, biomarkers, and results were recorded, as well as qualitative and quantitative definitions of EED. Biomarkers were organized into five EED domains, and the number of studies that support or do not support relationships among domains and between each domain with stunting were summarized., Results: There was little evidence to support the pathway from intestinal permeability to microbial translocation and from microbial translocation to stunting, but stronger support existed for the link between intestinal inflammation and systemic inflammation and for intestinal inflammation and stunting. There was conflicting evidence for the pathways from intestinal damage to intestinal permeability and intestinal damage to stunting., Conclusions: These results suggest that certain EED biomarkers may require reconsideration, particularly those most difficult to measure, such as microbial translocation and intestinal permeability. We discuss several issues with currently used biomarkers and recommend further analysis of pathogen-induced changes to the intestinal microbiota as a pathway leading to stunting.
- Published
- 2018
- Full Text
- View/download PDF
37. Antimicrobial Resistance Gene Acquisition and Depletion Following Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection.
- Author
-
Leung V, Vincent C, Edens TJ, Miller M, and Manges AR
- Subjects
- Anti-Bacterial Agents pharmacology, Feces microbiology, Genes, Bacterial, High-Throughput Nucleotide Sequencing, Humans, Recurrence, Clostridium Infections therapy, Drug Resistance, Multiple, Bacterial genetics, Fecal Microbiota Transplantation adverse effects, Gastrointestinal Microbiome, Genes, MDR
- Abstract
Fecal microbiota transplantation (FMT) may be a novel approach to eliminate multidrug-resistant bacteria from the gut and to prevent future infections. Using whole metagenome sequencing data from 8 FMT donor-recipient pairs, we identified 37 and 95 antimicrobial resistance genes that were acquired by or removed from FMT recipients, respectively., (© The Author(s) 2017. Published by Oxford University Press for the Infectious Diseases Society of America.)
- Published
- 2018
- Full Text
- View/download PDF
38. Risk factors for acquisition of multidrug-resistant Escherichia coli and development of community-acquired urinary tract infections.
- Author
-
Ukah UV, Glass M, Avery B, Daignault D, Mulvey MR, Reid-Smith RJ, Parmley EJ, Portt A, Boerlin P, and Manges AR
- Subjects
- Adult, Animals, Canada epidemiology, Case-Control Studies, Community-Acquired Infections epidemiology, Community-Acquired Infections microbiology, Enteropathogenic Escherichia coli drug effects, Escherichia coli Infections microbiology, Female, Humans, Incidence, Middle Aged, Poultry, Poultry Products, Risk Factors, Urinary Tract Infections microbiology, Young Adult, Drug Resistance, Multiple, Bacterial, Enteropathogenic Escherichia coli physiology, Escherichia coli Infections epidemiology, Urinary Tract Infections epidemiology
- Abstract
We examined risk factors associated with the intestinal acquisition of antimicrobial-resistant extraintestinal pathogenic Escherichia coli (ExPEC) and development of community-acquired urinary tract infection (UTI) in a case-control study of young women across Canada. A total of 399 women were recruited; 164 women had a UTI caused by E. coli resistant to ⩾1 antimicrobial classes and 98 had a UTI caused by E. coli resistant to ⩾3 antimicrobial classes. After adjustment for age, student health service (region of Canada) and either prior antibiotic use or UTI history, consumption of processed or ground chicken, cooked or raw shellfish, street foods and any organic fruit; as well as, contact with chickens, dogs and pet treats; and travel to Asia, were associated with an increased risk of UTI caused by antimicrobial resistant E. coli. A decreased risk of antimicrobial resistant UTI was associated with consumption of apples, nectarines, peppers, fresh herbs, peanuts and cooked beef. Drug-resistant UTI linked to foodborne and environmental exposures may be a significant public health concern and understanding the risk factors for intestinal acquisition of existing or newly emerging lineages of drug-resistant ExPEC is important for epidemiology, antimicrobial stewardship and prevention efforts.
- Published
- 2018
- Full Text
- View/download PDF
39. Integron-Associated DfrB4, a Previously Uncharacterized Member of the Trimethoprim-Resistant Dihydrofolate Reductase B Family, Is a Clinically Identified Emergent Source of Antibiotic Resistance.
- Author
-
Toulouse JL, Edens TJ, Alejaldre L, Manges AR, and Pelletier JN
- Subjects
- Drug Resistance, Bacterial genetics, Escherichia coli drug effects, Escherichia coli isolation & purification, Humans, Microbial Sensitivity Tests, Urinary Tract Infections drug therapy, Urinary Tract Infections microbiology, Anti-Infective Agents, Urinary pharmacology, Escherichia coli genetics, Escherichia coli Proteins genetics, Integrons genetics, Tetrahydrofolate Dehydrogenase genetics, Trimethoprim pharmacology, Trimethoprim Resistance genetics
- Abstract
Whole-genome sequencing of trimethoprim-resistant Escherichia coli clinical isolates identified a member of the trimethoprim-resistant type II dihydrofolate reductase gene family ( dfrB ). The dfrB4 gene was located within a class I integron flanked by multiple resistance genes. This arrangement was previously reported in a 130.6-kb multiresistance plasmid. The DfrB4 protein conferred a >2,000-fold increased trimethoprim resistance on overexpression in E. coli Our results are consistent with the finding that dfrB4 contributes to clinical trimethoprim resistance., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
40. Clonal distribution and associated characteristics of Escherichia coli clinical and surveillance isolates from a military medical center.
- Author
-
Manges AR, Mende K, Murray CK, Johnston BD, Sokurenko EV, Tchesnokova V, and Johnson JR
- Subjects
- Anti-Bacterial Agents therapeutic use, Drug Resistance, Microbial drug effects, Escherichia coli drug effects, Escherichia coli genetics, Escherichia coli Infections drug therapy, Hospitals, Military, Humans, Military Personnel, Molecular Epidemiology methods, Multilocus Sequence Typing methods, beta-Lactamases genetics, Escherichia coli isolation & purification, Escherichia coli Infections microbiology
- Abstract
Antimicrobial-resistant Escherichia coli are a concern for military health services. We studied 100 extended-spectrum beta-lactamase (ESBL)-producing and non-producing E. coli clinical and surveillance isolates from military personnel and civilians at Brooke Army Medical Center (2007-2011). Major E. coli lineages, most prominently ST10 (24%), ST131 (16%), and ST648 (8%), were distributed much as reported for other North American populations. ST131, represented mainly by its resistance-associated ST131-H30 clonal subset, was uniquely associated with a clinical origin, regardless of ESBL status. Thus, clonal background predicted resistance phenotype and clinical versus surveillance origin, and these findings could assist military clinicians and epidemiologists., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
41. From bugs to brains: The microbiome in neurological health.
- Author
-
McKay KA, Kowalec K, Brinkman F, Finlay BB, Horwitz M, Manges AR, Osborne L, and Tremlett H
- Subjects
- Animals, Brain physiopathology, British Columbia, Computational Biology methods, Gastrointestinal Microbiome genetics, Helminths, Humans, Multiple Sclerosis epidemiology, Multiple Sclerosis microbiology, Multiple Sclerosis virology, Brain physiology, Gastrointestinal Microbiome physiology
- Abstract
Knowledge surrounding the trillions of microbes that inhabit the human gut has bloomed exponentially in recent years, and the emerging concept of a gut-brain axis represents a major shift in how we think about neurological health. A recent workshop at the University of British Columbia, Canada brought together multi-disciplinary leaders in the field of microbiomics and brain health and aimed to serve as a springboard for future combined endeavors in these areas. This article provides the motivation for, and overview of, the workshop, and the next steps in establishing a cross-disciplinary initiative on Brain Health and the Microbiome., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
42. Linear growth trajectories in Zimbabwean infants.
- Author
-
Gough EK, Moodie EE, Prendergast AJ, Ntozini R, Moulton LH, Humphrey JH, and Manges AR
- Subjects
- Birth Weight, Body Height, Child, Preschool, Cluster Analysis, Cohort Studies, Female, HIV Infections, Humans, Infant, Infant Nutritional Physiological Phenomena, Logistic Models, Longitudinal Studies, Male, Nutritional Status, Socioeconomic Factors, Vitamin A administration & dosage, Zimbabwe, Child Development, Growth Disorders epidemiology, Malnutrition epidemiology
- Abstract
Background: Undernutrition in early life underlies 45% of child deaths globally. Stunting malnutrition (suboptimal linear growth) also has long-term negative effects on childhood development. Linear growth deficits accrue in the first 1000 d of life. Understanding the patterns and timing of linear growth faltering or recovery during this period is critical to inform interventions to improve infant nutritional status., Objective: We aimed to identify the pattern and determinants of linear growth trajectories from birth through 24 mo of age in a cohort of Zimbabwean infants., Design: We performed a secondary analysis of longitudinal data from a subset of 3338 HIV-unexposed infants in the Zimbabwe Vitamin A for Mothers and Babies trial. We used k-means clustering for longitudinal data to identify linear growth trajectories and multinomial logistic regression to identify covariates that were associated with each trajectory group., Results: For the entire population, the mean length-for-age z score declined from -0.6 to -1.4 between birth and 24 mo of age. Within the population, 4 growth patterns were identified that were each characterized by worsening linear growth restriction but varied in the timing and severity of growth declines. In our multivariable model, 1-U increments in maternal height and education and infant birth weight and length were associated with greater relative odds of membership in the least-growth restricted groups (A and B) and reduced odds of membership in the more-growth restricted groups (C and D). Male infant sex was associated with reduced odds of membership in groups A and B but with increased odds of membership in groups C and D., Conclusion: In this population, all children were experiencing growth restriction but differences in magnitude were influenced by maternal height and education and infant sex, birth weight, and birth length, which suggest that key determinants of linear growth may already be established by the time of birth. This trial was registered at clinicaltrials.gov as NCT00198718.
- Published
- 2016
- Full Text
- View/download PDF
43. Fecal microbiota transplantation for the intestinal decolonization of extensively antimicrobial-resistant opportunistic pathogens: a review.
- Author
-
Manges AR, Steiner TS, and Wright AJ
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Bacteria drug effects, Bacteria pathogenicity, Bacterial Infections microbiology, Female, Gastrointestinal Diseases microbiology, Gastrointestinal Microbiome physiology, Humans, Immunocompromised Host, Male, Middle Aged, Opportunistic Infections microbiology, Bacterial Infections therapy, Drug Resistance, Multiple, Bacterial, Fecal Microbiota Transplantation, Gastrointestinal Diseases therapy, Opportunistic Infections therapy
- Abstract
Treatment options for multidrug-resistant (MDR) bacterial infections are limited and often less effective. Non-pharmacologic approaches to preventing or treating MDR infections are currently restricted to improved antimicrobial stewardship and infection control practices. Fecal microbiota transplantation (FMT), a highly effective treatment for recurrent Clostridium difficile infection, has emerged as a promising therapy for intestinal MDR bacterial decolonization. A total of eight case reports have been published showing FMT resulted in intestinal decolonization of extended spectrum β-lactamase (ESBL)-producing and carbapenemase-producing Enterobacteriaceae, vancomycin-resistant Enterococci, or methicillin-resistant Staphylococcus aureus. The procedure has been shown to work even in immunocompromised patients and those experiencing medical crises without any adverse events. Five trials are currently underway to further investigate the use of FMT for MDR bacterial decolonization. FMT is a completely novel way to eradicate drug-resistant bacteria from the intestinal reservoir and should be further investigated to address the global problem of difficult-to-treat, MDR bacterial infections.
- Published
- 2016
- Full Text
- View/download PDF
44. Evolutionary History of the Global Emergence of the Escherichia coli Epidemic Clone ST131.
- Author
-
Stoesser N, Sheppard AE, Pankhurst L, De Maio N, Moore CE, Sebra R, Turner P, Anson LW, Kasarskis A, Batty EM, Kos V, Wilson DJ, Phetsouvanh R, Wyllie D, Sokurenko E, Manges AR, Johnson TJ, Price LB, Peto TE, Johnson JR, Didelot X, Walker AS, and Crook DW
- Subjects
- Chromosomes, Bacterial, Drug Resistance, Bacterial, Escherichia coli classification, Escherichia coli isolation & purification, Genes, Bacterial, Global Health, Humans, Molecular Epidemiology, Plasmids, Sequence Analysis, DNA, Epidemics, Escherichia coli genetics, Escherichia coli Infections epidemiology, Escherichia coli Infections microbiology, Evolution, Molecular, Genotype
- Abstract
Unlabelled: Escherichia colisequence type 131 (ST131) has emerged globally as the most predominant extraintestinal pathogenic lineage within this clinically important species, and its association with fluoroquinolone and extended-spectrum cephalosporin resistance impacts significantly on treatment. The evolutionary histories of this lineage, and of important antimicrobial resistance elements within it, remain unclearly defined. This study of the largest worldwide collection (n= 215) of sequenced ST131E. coliisolates to date demonstrates that the clonal expansion of two previously recognized antimicrobial-resistant clades, C1/H30R and C2/H30Rx, started around 25 years ago, consistent with the widespread introduction of fluoroquinolones and extended-spectrum cephalosporins in clinical medicine. These two clades appear to have emerged in the United States, with the expansion of the C2/H30Rx clade driven by the acquisition of ablaCTX-M-15-containing IncFII-like plasmid that has subsequently undergone extensive rearrangement. Several other evolutionary processes influencing the trajectory of this drug-resistant lineage are described, including sporadic acquisitions of CTX-M resistance plasmids and chromosomal integration ofblaCTX-Mwithin subclusters followed by vertical evolution. These processes are also occurring for another family of CTX-M gene variants more recently observed among ST131, theblaCTX-M-14/14-likegroup. The complexity of the evolutionary history of ST131 has important implications for antimicrobial resistance surveillance, epidemiological analysis, and control of emerging clinical lineages ofE. coli These data also highlight the global imperative to reduce specific antibiotic selection pressures and demonstrate the important and varied roles played by plasmids and other mobile genetic elements in the perpetuation of antimicrobial resistance within lineages., Importance: Escherichia coli, perennially a major bacterial pathogen, is becoming increasingly difficult to manage due to emerging resistance to all preferred antimicrobials. Resistance is concentrated within specificE. colilineages, such as sequence type 131 (ST131). Clarification of the genetic basis for clonally associated resistance is key to devising intervention strategies. We used high-resolution genomic analysis of a large global collection of ST131 isolates to define the evolutionary history of extended-spectrum beta-lactamase production in ST131. We documented diverse contributory genetic processes, including stable chromosomal integrations of resistance genes, persistence and evolution of mobile resistance elements within sublineages, and sporadic acquisition of different resistance elements. Both global distribution and regional segregation were evident. The diversity of resistance element acquisition and propagation within ST131 indicates a need for control and surveillance strategies that target both bacterial strains and mobile genetic elements., (Copyright © 2016 Stoesser et al.)
- Published
- 2016
- Full Text
- View/download PDF
45. Bloom and bust: intestinal microbiota dynamics in response to hospital exposures and Clostridium difficile colonization or infection.
- Author
-
Vincent C, Miller MA, Edens TJ, Mehrotra S, Dewar K, and Manges AR
- Subjects
- Aged, Bile Acids and Salts biosynthesis, Cephalosporins adverse effects, Clostridioides difficile drug effects, Clostridioides difficile growth & development, Cross Infection pathology, Diarrhea etiology, Diarrhea pathology, Enterocolitis, Pseudomembranous etiology, Enterocolitis, Pseudomembranous pathology, Eubacterium drug effects, Eubacterium growth & development, Eubacterium pathogenicity, Female, Fluoroquinolones adverse effects, Humans, Laxatives adverse effects, Male, Middle Aged, Prospective Studies, Anti-Bacterial Agents adverse effects, Clostridioides difficile pathogenicity, Cross Infection microbiology, Diarrhea microbiology, Enterocolitis, Pseudomembranous microbiology, Gastrointestinal Microbiome genetics, Metagenome
- Abstract
Background: Clostridium difficile infection (CDI) is the leading infectious cause of nosocomial diarrhea. Hospitalized patients are at increased risk of developing CDI because they are exposed to C. difficile spores through contact with the hospital environment and often receive antibiotics and other medications that can disrupt the integrity of the indigenous intestinal microbiota and impair colonization resistance. Using whole metagenome shotgun sequencing, we examined the diversity and composition of the fecal microbiota in a prospective cohort study of 98 hospitalized patients., Results: Four patients had asymptomatic C. difficile colonization, and four patients developed CDI. We observed dramatic shifts in the structure of the gut microbiota during hospitalization. In contrast to CDI cases, asymptomatic patients exhibited elevated relative abundance of potentially protective bacterial taxa in their gut at the onset of C. difficile colonization. Use of laxatives was associated with significant reductions in the relative abundance of Clostridium and Eubacterium; species within these genera have previously been shown to enhance resistance to CDI via the production of secondary bile acids. Cephalosporin and fluoroquinolone exposure decreased the frequency of Clostridiales Family XI Incertae Sedis, a bacterial family that has been previously associated with decreased CDI risk., Conclusions: This study underscores the detrimental impact of antibiotics as well as other medications, particularly laxatives, on the intestinal microbiota and suggests that co-colonization with key bacterial taxa may prevent C. difficile overgrowth or the transition from asymptomatic C. difficile colonization to CDI.
- Published
- 2016
- Full Text
- View/download PDF
46. Escherichia coli and urinary tract infections: the role of poultry-meat.
- Author
-
Manges AR
- Subjects
- Animals, Drug Resistance, Multiple, Bacterial, Escherichia coli genetics, Escherichia coli Infections microbiology, Escherichia coli Infections veterinary, Food Microbiology, Humans, Phylogeny, Poultry Diseases microbiology, Poultry Diseases transmission, Zoonoses microbiology, Escherichia coli classification, Escherichia coli Infections transmission, Meat microbiology, Poultry microbiology, Urinary Tract Infections microbiology
- Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is the most common cause of community-acquired and hospital-acquired extraintestinal infections. The hypothesis that human ExPEC may have a food animal reservoir has been a topic of investigation by multiple groups around the world. Experimental studies showing the shared pathogenic potential of human ExPEC and avian pathogenic E. coli suggest that these extraintestinal E. coli may be derived from the same bacterial lineages or share common evolutionary roots. The consistent observation of specific human ExPEC lineages in poultry or poultry products, and rarely in other meat commodities, supports the hypothesis that there may be a poultry reservoir for human ExPEC. The time lag between human ExPEC acquisition (in the intestine) and infection is the fundamental challenge facing studies attempting to attribute ExPEC transmission to poultry or other environmental sources. Even whole genome sequencing efforts to address attribution will struggle with defining meaningful genetic relationships outside of a discrete food-borne outbreak setting. However, if even a fraction of all human ExPEC infections, especially antimicrobial-resistant ExPEC infections, is attributable to the introduction of multidrug-resistant ExPEC lineages through contaminated food product(s), the relevance to public health, food animal production and food safety will be significant., (Copyright © 2016 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. Erratum to: Linear growth faltering in infants is associated with Acidaminococcus sp. and community-level changes in the gut microbiota.
- Author
-
Gough EK, Stephens DA, Moodie EE, Prendergast AJ, Stoltzfus RJ, Humphrey JH, and Manges AR
- Published
- 2016
- Full Text
- View/download PDF
48. Assessing the Intestinal Microbiota in the SHINE Trial.
- Author
-
Gough EK, Prendergast AJ, Mutasa KE, Stoltzfus RJ, and Manges AR
- Subjects
- Diet, Female, Gastrointestinal Microbiome genetics, Humans, Infant, Intestines microbiology, Male, Molecular Typing, Randomized Controlled Trials as Topic methods, Research Design, Rural Population, Sequence Analysis, DNA, Zimbabwe, Gastrointestinal Microbiome physiology, Growth Disorders physiopathology, Infant Nutritional Physiological Phenomena, Intestines physiopathology
- Abstract
Advances in DNA sequencing technology now allow us to explore the dynamics and functions of the microbes that inhabit the human body, the microbiota. Recent studies involving experimental animal models suggest a role of the gut microbiota in growth. However, the specific changes in the human gut microbiota that contribute to growth remain unclear, and studies investigating the gut microbiota as a determinant of environmental enteric dysfunction (EED) and child stunting are lacking. In this article, we review the evidence for a link between the developing infant gut microbiota, infant feeding, EED, and stunting, and discuss the potential causal pathways relating these variables. We outline the analytic approaches we will use to investigate these relationships, by capitalizing on the longitudinal design and randomized interventions of the Sanitation Hygiene Infant Nutrition Efficacy trial in Zimbabwe., (© The Author 2015. Published by Oxford University Press for the Infectious Diseases Society of America.)
- Published
- 2015
- Full Text
- View/download PDF
49. Reservoirs of Extraintestinal Pathogenic Escherichia coli.
- Author
-
Manges AR and Johnson JR
- Subjects
- Animals, Escherichia coli Infections microbiology, Foodborne Diseases epidemiology, Foodborne Diseases microbiology, Humans, Disease Reservoirs, Environmental Exposure, Escherichia coli Infections epidemiology, Escherichia coli Infections transmission, Meat Products microbiology, Pets
- Abstract
Several potential reservoirs for the Escherichia coli strains that cause most human extraintestinal infections (extraintestinal pathogenic E. coli; ExPEC) have been identified, including the human intestinal tract and various non-human reservoirs, such as companion animals, food animals, retail meat products, sewage, and other environmental sources. Understanding ExPEC reservoirs, chains of transmission, transmission dynamics, and epidemiologic associations will assist greatly in finding ways to reduce the ExPEC-associated disease burden. The need to clarify the ecological behavior of ExPEC is all the more urgent because environmental reservoirs may contribute to acquisition of antimicrobial resistance determinants and selection for and amplification of resistant ExPEC. In this chapter, we review the evidence for different ExPEC reservoirs, with particular attention to food and food animals, and discuss the public health implications of these reservoirs for ExPEC dissemination and transmission.
- Published
- 2015
- Full Text
- View/download PDF
50. Editorial Commentary: Genomic Epidemiology: Revealing Hidden Reservoirs for Klebsiella pneumoniae.
- Author
-
Manges AR
- Subjects
- Animals, Humans, Klebsiella Infections microbiology, Klebsiella pneumoniae classification, Klebsiella pneumoniae isolation & purification, Meat microbiology, Urinary Tract Infections microbiology
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.