Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Microbiol Rev ; 36(1): e0011922, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36847515

RESUMEN

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.


Asunto(s)
Virus , Virus/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Bacterias/genética , Metagenómica/métodos , Genoma Viral/genética
2.
J Infect Dis ; 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38696336

RESUMEN

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.

3.
Clin Microbiol Rev ; 32(3)2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31189557

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Escherichia coli Patógena Extraintestinal/genética , Farmacorresistencia Bacteriana , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/prevención & control , Escherichia coli Patógena Extraintestinal/clasificación , Escherichia coli Patógena Extraintestinal/patogenicidad , Genoma Bacteriano/genética , Humanos
4.
BMC Med ; 18(1): 392, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33317529

RESUMEN

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.


Asunto(s)
Lactancia Materna/métodos , Farmacorresistencia Bacteriana/fisiología , Microbioma Gastrointestinal/inmunología , Leche Humana/microbiología , Femenino , Humanos , Recién Nacido , Masculino
5.
Clin Infect Dis ; 66(3): 456-457, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29020222

RESUMEN

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.


Asunto(s)
Infecciones por Clostridium/terapia , Farmacorresistencia Bacteriana Múltiple/genética , Trasplante de Microbiota Fecal/efectos adversos , Microbioma Gastrointestinal , Genes MDR , Antibacterianos/farmacología , Heces/microbiología , Genes Bacterianos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Recurrencia
6.
Epidemiol Infect ; 147: e55, 2018 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-30501663

RESUMEN

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.

7.
Artículo en Inglés | MEDLINE | ID: mdl-28242670

RESUMEN

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.


Asunto(s)
Antiinfecciosos Urinarios/farmacología , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Integrones/genética , Tetrahidrofolato Deshidrogenasa/genética , Resistencia al Trimetoprim/genética , Trimetoprim/farmacología , Farmacorresistencia Bacteriana/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/aislamiento & purificación , Humanos , Pruebas de Sensibilidad Microbiana , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología
8.
Clin Infect Dis ; 61 Suppl 7: S738-44, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26602302

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Trastornos del Crecimiento/fisiopatología , Fenómenos Fisiológicos Nutricionales del Lactante , Intestinos/fisiopatología , Dieta , Femenino , Microbioma Gastrointestinal/genética , Humanos , Lactante , Intestinos/microbiología , Masculino , Tipificación Molecular , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Proyectos de Investigación , Población Rural , Análisis de Secuencia de ADN , Zimbabwe
9.
Foodborne Pathog Dis ; 12(4): 302-10, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25774654

RESUMEN

We investigated whether specific sequence types, and their shared virulence gene profiles, may be associated with both human and food animal reservoirs. A total of 600 Escherichia coli isolates were assembled from human (n=265) and food-animal (n=335) sources from overlapping geographic areas and time periods (2005-2010) in Canada. The entire collection was subjected to multilocus sequence typing and a subset of 286 E. coli isolates was subjected to an E. coli-specific virulence gene microarray. The most common sequence type (ST) was E. coli ST10, which was present in all human and food-animal sources, followed by ST69, ST73, ST95, ST117, and ST131. A core group of virulence genes was associated with all 10 common STs including artJ, ycfZ, csgA, csgE, fimA, fimH, gad, hlyE, ibeB, mviM, mviN, and ompA. STs 73, 92, and 95 exhibited the largest number of virulence genes, and all were exclusively identified from human infections. ST117 was found in both human and food-animal sources and shared virulence genes common in extraintestinal pathogenic E. coli lineages. Select groups of E. coli may be found in both human and food-animal reservoirs.


Asunto(s)
Proteínas de Escherichia coli/genética , Escherichia coli/clasificación , Escherichia coli/aislamiento & purificación , Carne/microbiología , Tipificación de Secuencias Multilocus , Animales , Técnicas de Tipificación Bacteriana , Canadá , Infecciones por Escherichia coli/microbiología , Genes Bacterianos , Humanos , Factores de Virulencia/genética
10.
Lancet Reg Health Am ; 29: 100658, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38235369

RESUMEN

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).

11.
Artículo en Inglés | MEDLINE | ID: mdl-38616557

RESUMEN

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.

12.
Urolithiasis ; 52(1): 38, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38413462

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Cálculos Renales , Humanos , Oxalatos/metabolismo , Factores de Riesgo , Bacterias/genética , Butiratos , Cálculos Renales/microbiología
13.
medRxiv ; 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38293149

RESUMEN

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.

15.
Appl Environ Microbiol ; 79(12): 3892-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23584785

RESUMEN

This study identified and characterized enteropathogenic Escherichia coli (EPEC) in the Canadian food supply. Eighteen of 450 E. coli isolates from food animal sources were identified as atypical EPEC (aEPEC). Several of the aEPEC isolates identified in this study possessed multiple virulence genes, exhibited adherence and attaching and effacing (A/E) lesion formation, disrupted tight junctions, and were coclassified with the extraintestinal pathogenic E. coli (ExPEC) and enterotoxigenic E. coli (ETEC) pathotypes.


Asunto(s)
Enfermedades de los Bovinos/epidemiología , Pollos , Escherichia coli Enteropatógena/genética , Infecciones por Escherichia coli/veterinaria , Carne/microbiología , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de los Porcinos/epidemiología , Mataderos/normas , Animales , Western Blotting/veterinaria , Canadá/epidemiología , Bovinos , Enfermedades de los Bovinos/microbiología , Análisis por Conglomerados , Farmacorresistencia Microbiana/genética , Infecciones por Escherichia coli/epidemiología , Abastecimiento de Alimentos/normas , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa/veterinaria , Enfermedades de las Aves de Corral/microbiología , Porcinos , Enfermedades de los Porcinos/microbiología
16.
Int J Antimicrob Agents ; 61(1): 106690, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36372343

RESUMEN

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.


Asunto(s)
Infecciones por VIH , Desnutrición Aguda Severa , Humanos , Niño , Lactante , Antibacterianos/uso terapéutico , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Desnutrición Aguda Severa/complicaciones , Desnutrición Aguda Severa/terapia
17.
Nat Commun ; 14(1): 654, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36788215

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Infecciones por VIH , Lactante , Niño , Humanos , Preescolar , Microbioma Gastrointestinal/genética , Prevalencia , Trastornos del Crecimiento/epidemiología , Abastecimiento de Agua
18.
Front Cell Infect Microbiol ; 13: 1144254, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37065202

RESUMEN

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 cpn60-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.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Recién Nacido , Humanos , Lactante , Embarazo , Femenino , Microbioma Gastrointestinal/genética , Estudios Prospectivos , Canadá , Heces/microbiología
19.
Clin Infect Dis ; 55(5): 712-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22615330

RESUMEN

Most human extraintestinal Escherichia coli infections, including those involving antimicrobial resistant strains, are caused by the members of a limited number of distinctive E. coli lineages, termed extraintestinal pathogenic E. coli (ExPEC), that have a special ability to cause disease at extraintestinal sites when they exit their usual reservoir in the host's intestinal tract. Multiple lines of evidence suggest that many of the ExPEC strains encountered in humans with urinary tract infection, sepsis, and other extraintestinal infections, especially the most extensively antimicrobial-resistant strains, may have a food animal source, and may be transmitted to humans via the food supply. This review summarizes the evidence that food-borne organisms are a significant cause of extraintestinal E. coli infections in humans.


Asunto(s)
Deficiencias de Hierro , Malaria Falciparum/epidemiología , Femenino , Humanos , Masculino
20.
Emerg Infect Dis ; 18(3): 415-21, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22377351

RESUMEN

We previously described how retail meat, particularly chicken, might be a reservoir for extraintestinal pathogenic Escherichia coli (ExPEC) causing urinary tract infections (UTIs) in humans. To rule out retail beef and pork as potential reservoirs, we tested 320 additional E. coli isolates from these meats. Isolates from beef and pork were significantly less likely than those from chicken to be genetically related to isolates from humans with UTIs. We then tested whether the reservoir for ExPEC in humans could be food animals themselves by comparing geographically and temporally matched E. coli isolates from 475 humans with UTIs and from cecal contents of 349 slaughtered animals. We found genetic similarities between E. coli from animals in abattoirs, principally chickens, and ExPEC causing UTIs in humans. ExPEC transmission from food animals could be responsible for human infections, and chickens are the most probable reservoir.


Asunto(s)
Reservorios de Enfermedades , Infecciones por Escherichia coli/transmisión , Escherichia coli/aislamiento & purificación , Carne/microbiología , Infecciones Urinarias/microbiología , Infecciones Urinarias/transmisión , Adolescente , Adulto , Animales , Canadá/epidemiología , Bovinos , Pollos , Infecciones por Escherichia coli/epidemiología , Femenino , Microbiología de Alimentos , Humanos , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Porcinos , Infecciones Urinarias/epidemiología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA