Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Clin Microbiol ; 57(9)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315955

RESUMEN

We applied metagenomic next-generation sequencing (mNGS) to detect Zaire Ebola virus (EBOV) and other potential pathogens from whole-blood samples from 70 patients with suspected Ebola hemorrhagic fever during a 2014 outbreak in Boende, Democratic Republic of the Congo (DRC) and correlated these findings with clinical symptoms. Twenty of 31 patients (64.5%) tested in Kinshasa, DRC, were EBOV positive by quantitative reverse transcriptase PCR (qRT-PCR). Despite partial degradation of sample RNA during shipping and handling, mNGS followed by EBOV-specific capture probe enrichment in a U.S. genomics laboratory identified EBOV reads in 22 of 70 samples (31.4%) versus in 21 of 70 (30.0%) EBOV-positive samples by repeat qRT-PCR (overall concordance = 87.1%). Reads from Plasmodium falciparum (malaria) were detected in 21 patients, of which at least 9 (42.9%) were coinfected with EBOV. Other positive viral detections included hepatitis B virus (n = 2), human pegivirus 1 (n = 2), Epstein-Barr virus (n = 9), and Orungo virus (n = 1), a virus in the Reoviridae family. The patient with Orungo virus infection presented with an acute febrile illness and died rapidly from massive hemorrhage and dehydration. Although the patient's blood sample was negative by EBOV qRT-PCR testing, identification of viral reads by mNGS confirmed the presence of EBOV coinfection. In total, 9 new EBOV genomes (3 complete genomes, and an additional 6 ≥50% complete) were assembled. Relaxed molecular clock phylogenetic analysis demonstrated a molecular evolutionary rate for the Boende strain 4 to 10× slower than that of other Ebola lineages. These results demonstrate the utility of mNGS in broad-based pathogen detection and outbreak surveillance.


Asunto(s)
Coinfección/epidemiología , Brotes de Enfermedades , Ebolavirus/clasificación , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/virología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Metagenómica/métodos , Adulto , Coinfección/parasitología , Coinfección/patología , Coinfección/virología , República Democrática del Congo/epidemiología , Ebolavirus/genética , Ebolavirus/aislamiento & purificación , Femenino , Fiebre Hemorrágica Ebola/parasitología , Fiebre Hemorrágica Ebola/patología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
2.
Clin Infect Dis ; 65(9): 1477-1485, 2017 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-29020199

RESUMEN

BACKGROUND: Twelve percent of all acute liver failure (ALF) cases are of unknown origin, often termed indeterminate. A previously unrecognized hepatotropic virus has been suspected as a potential etiologic agent. METHODS: We compared the performance of metagenomic next-generation sequencing (mNGS) with confirmatory nucleic acid testing (NAT) to routine clinical diagnostic testing in detection of known or novel viruses associated with ALF. Serum samples from 204 adult ALF patients collected from 1998 to 2010 as part of a nationwide registry were analyzed. One hundred eighty-seven patients (92%) were classified as indeterminate, while the remaining 17 patients (8%) served as controls, with infections by either hepatitis A virus or hepatitis B virus (HBV), or a noninfectious cause for their ALF. RESULTS: Eight cases of infection from previously unrecognized viral pathogens were detected by mNGS (4 cases of herpes simplex virus type 1, including 1 case of coinfection with HBV, and 1 case each of HBV, parvovirus B19, cytomegalovirus, and human herpesvirus 7). Several missed dual or triple infections were also identified, and assembled viral genomes provided additional information on genotyping and drug resistance mutations. Importantly, no sequences corresponding to novel viruses were detected. CONCLUSIONS: These results suggest that ALF patients should be screened for the presence of uncommon viruses and coinfections, and that most cases of indeterminate ALF in the United States do not appear to be caused by novel viral pathogens. In the future, mNGS testing may be useful for comprehensive diagnosis of viruses associated with ALF, or to exclude infectious etiologies.


Asunto(s)
Genoma Viral/genética , Virus de Hepatitis/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Fallo Hepático Agudo/virología , Metagenómica/métodos , Adolescente , Adulto , ADN Viral/genética , Femenino , Hepatitis Viral Humana/diagnóstico , Hepatitis Viral Humana/genética , Humanos , Masculino , Persona de Mediana Edad , ARN Viral/genética , Estudios Retrospectivos , Adulto Joven
3.
Emerg Infect Dis ; 23(10): 1964-1968, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28930022
4.
N Engl J Med ; 370(25): 2408-17, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24896819

RESUMEN

A 14-year-old boy with severe combined immunodeficiency presented three times to a medical facility over a period of 4 months with fever and headache that progressed to hydrocephalus and status epilepticus necessitating a medically induced coma. Diagnostic workup including brain biopsy was unrevealing. Unbiased next-generation sequencing of the cerebrospinal fluid identified 475 of 3,063,784 sequence reads (0.016%) corresponding to leptospira infection. Clinical assays for leptospirosis were negative. Targeted antimicrobial agents were administered, and the patient was discharged home 32 days later with a status close to his premorbid condition. Polymerase-chain-reaction (PCR) and serologic testing at the Centers for Disease Control and Prevention (CDC) subsequently confirmed evidence of Leptospira santarosai infection.


Asunto(s)
Encéfalo/patología , Líquido Cefalorraquídeo/microbiología , ADN Bacteriano/análisis , Leptospira/genética , Leptospirosis/diagnóstico , Meningoencefalitis/diagnóstico , Análisis de Secuencia de ADN/métodos , Adenosina Desaminasa/deficiencia , Adolescente , Agammaglobulinemia/complicaciones , Biopsia , Fiebre/etiología , Cefalea/etiología , Humanos , Leptospira/aislamiento & purificación , Leptospirosis/complicaciones , Leptospirosis/microbiología , Masculino , Meningoencefalitis/complicaciones , Meningoencefalitis/microbiología , Inmunodeficiencia Combinada Grave/complicaciones
5.
Emerg Infect Dis ; 22(10): 1788-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27448188

RESUMEN

Sequencing of isolates from patients in Bahia, Brazil, where most Zika virus cases in Brazil have been reported, resulted in 11 whole and partial Zika virus genomes. Phylogenetic analyses revealed a well-supported Bahia-specific Zika virus lineage, which indicates sustained Zika virus circulation in Salvador, Bahia's capital city, since mid-2014.


Asunto(s)
Infección por el Virus Zika/virología , Virus Zika/clasificación , Adulto , Anciano , Brasil/epidemiología , ADN Viral , Femenino , Genoma Viral , Humanos , Masculino , Tipificación Molecular , Filogenia , Análisis de Secuencia de ADN , Virus Zika/genética , Infección por el Virus Zika/epidemiología
6.
J Clin Microbiol ; 54(9): 2348-53, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27413190

RESUMEN

Metagenomic next-generation sequencing (mNGS) of samples from 15 patients with documented Zika virus (ZIKV) infection in Bahia, Brazil, from April 2015 to January 2016 identified coinfections with chikungunya virus (CHIKV) in 2 of 15 ZIKV-positive cases by PCR (13.3%). While generally nonspecific, the clinical presentation corresponding to these two CHIKV/ZIKV coinfections reflected infection by the virus present at a higher titer. Aside from CHIKV and ZIKV, coinfections of other viral pathogens were not detected. The mNGS approach is promising for differential diagnosis of acute febrile illness and identification of coinfections, although targeted arbovirus screening may be sufficient in the current ZIKV outbreak setting.


Asunto(s)
Fiebre Chikungunya/epidemiología , Virus Chikungunya/aislamiento & purificación , Coinfección/epidemiología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Metagenómica/métodos , Infección por el Virus Zika/epidemiología , Virus Zika/aislamiento & purificación , Adulto , Brasil/epidemiología , Fiebre Chikungunya/diagnóstico , Fiebre Chikungunya/virología , Virus Chikungunya/genética , Coinfección/diagnóstico , Coinfección/virología , Femenino , Humanos , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular/métodos , Virus Zika/genética , Infección por el Virus Zika/diagnóstico , Infección por el Virus Zika/virología
7.
Commun Biol ; 4(1): 475, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846513

RESUMEN

COVID-19 is a respiratory illness caused by a novel coronavirus called SARS-CoV-2. The viral spike (S) protein engages the human angiotensin-converting enzyme 2 (ACE2) receptor to invade host cells with ~10-15-fold higher affinity compared to SARS-CoV S-protein, making it highly infectious. Here, we assessed if ACE2 polymorphisms can alter host susceptibility to SARS-CoV-2 by affecting this interaction. We analyzed over 290,000 samples representing >400 population groups from public genomic datasets and identified multiple ACE2 protein-altering variants. Using reported structural data, we identified natural ACE2 variants that could potentially affect virus-host interaction and thereby alter host susceptibility. These include variants S19P, I21V, E23K, K26R, T27A, N64K, T92I, Q102P and H378R that were predicted to increase susceptibility, while variants K31R, N33I, H34R, E35K, E37K, D38V, Y50F, N51S, M62V, K68E, F72V, Y83H, G326E, G352V, D355N, Q388L and D509Y were predicted to be protective variants that show decreased binding to S-protein. Using biochemical assays, we confirmed that K31R and E37K had decreased affinity, and K26R and T92I variants showed increased affinity for S-protein when compared to wildtype ACE2. Consistent with this, soluble ACE2 K26R and T92I were more effective in blocking entry of S-protein pseudotyped virus suggesting that ACE2 variants can modulate susceptibility to SARS-CoV-2.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/genética , Predisposición Genética a la Enfermedad/genética , Mutación Missense/genética , Polimorfismo Genético , Receptores Virales/genética , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , COVID-19/virología , Interacciones Huésped-Patógeno , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Receptores Virales/química , Receptores Virales/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
9.
Nat Microbiol ; 5(3): 443-454, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31932713

RESUMEN

Metagenomic next-generation sequencing (mNGS), the shotgun sequencing of RNA and DNA from clinical samples, has proved useful for broad-spectrum pathogen detection and the genomic surveillance of viral outbreaks. An additional target enrichment step is generally needed for high-sensitivity pathogen identification in low-titre infections, yet available methods using PCR or capture probes can be limited by high cost, narrow scope of detection, lengthy protocols and/or cross-contamination. Here, we developed metagenomic sequencing with spiked primer enrichment (MSSPE), a method for enriching targeted RNA viral sequences while simultaneously retaining metagenomic sensitivity for other pathogens. We evaluated MSSPE for 14 different viruses, yielding a median tenfold enrichment and mean 47% (±16%) increase in the breadth of genome coverage over mNGS alone. Virus detection using MSSPE arboviral or haemorrhagic fever viral panels was comparable in sensitivity to specific PCR, demonstrating 95% accuracy for the detection of Zika, Ebola, dengue, chikungunya and yellow fever viruses in plasma samples from infected patients. Notably, sequences from re-emerging and/or co-infecting viruses that have not been specifically targeted a priori, including Powassan and Usutu, were successfully enriched using MSSPE. MSSPE is simple, low cost, fast and deployable on either benchtop or portable nanopore sequencers, making this method directly applicable for diagnostic laboratory and field use.


Asunto(s)
Genoma Viral , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Metagenoma , Metagenómica/métodos , Virus/genética , Virus/aislamiento & purificación , Virus Chikungunya/genética , Virus Chikungunya/aislamiento & purificación , Biología Computacional , ADN Viral/genética , Dengue/diagnóstico , Virus del Dengue/genética , Virus del Dengue/aislamiento & purificación , Ebolavirus/genética , Ebolavirus/aislamiento & purificación , Fiebre Hemorrágica Ebola/diagnóstico , Humanos , Reacción en Cadena de la Polimerasa , ARN Viral/genética , ARN Viral/aislamiento & purificación , Virosis/diagnóstico , Fiebre Amarilla/diagnóstico , Virus Zika/genética , Infección por el Virus Zika/diagnóstico
10.
Cell Host Microbe ; 23(6): 855-864.e7, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29805095

RESUMEN

The Zika virus (ZIKV) epidemic in the Americas established ZIKV as a major public health threat and uncovered its association with severe diseases, including microcephaly. However, genetic epidemiology in some at-risk regions, particularly Central America and Mexico, remains limited. We report 61 ZIKV genomes from this region, generated using metagenomic sequencing with ZIKV-specific enrichment, and combine phylogenetic, epidemiological, and environmental data to reconstruct ZIKV transmission. These analyses revealed multiple independent ZIKV introductions to Central America and Mexico. One introduction, likely from Brazil via Honduras, led to most infections and the undetected spread of ZIKV through the region from late 2014. Multiple lines of evidence indicate biannual peaks of ZIKV transmission in the region, likely driven by varying local environmental conditions for mosquito vectors and herd immunity. The spatial and temporal heterogeneity of ZIKV transmission in Central America and Mexico challenges arbovirus surveillance and disease control measures.


Asunto(s)
Genoma Viral/genética , Mosquitos Vectores/virología , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/transmisión , Virus Zika/genética , Adolescente , Adulto , Brasil/epidemiología , América Central/epidemiología , Niño , Preescolar , Humanos , Inmunidad Colectiva/inmunología , Metagenómica , México/epidemiología , Filogenia , Análisis de Secuencia de ARN , Virus Zika/inmunología , Infección por el Virus Zika/sangre , Infección por el Virus Zika/orina
11.
Sci Rep ; 7(1): 17126, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29215081

RESUMEN

A monkey model of Zika virus (ZIKV) infection is urgently needed to better understand transmission and pathogenesis, given its proven association with fetal brain defects in pregnant women and acute neurological illness. Here we experimentally infected 4 male marmosets with ZIKV (prototype 1947 African strain) and monitored them clinically with sampling of various body fluids and tissues for nearly 3 months. We show that the course of acute infection with ZIKV in these New World monkeys resembles the human illness in many respects, including (1) lack of apparent clinical symptoms in most cases, (2) persistence of the virus in body fluids such as semen and saliva for longer periods of time than in serum, and (3) generation of neutralizing antibodies as well as an antiviral immunological host response. Importantly, ZIKV-infected saliva samples (in addition to serum) were found to be infectious, suggesting potential capacity for viral transmission by the oral route. Re-challenge of a previously infected marmoset with a contemporary outbreak strain SPH2015 from Brazil resulted in continued protection against infection, no viral shedding, and boosting of the immune response. Given the key similarities to human infection, a marmoset model of ZIKV infection may be useful for testing of new drugs and vaccines.


Asunto(s)
Infección por el Virus Zika/patología , Animales , Callithrix , Chlorocebus aethiops , Modelos Animales de Enfermedad , Masculino , Saliva/virología , Células Vero , Virus Zika/patogenicidad , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología
12.
Sci Rep ; 7(1): 18022, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29269933

RESUMEN

We evaluated the performance of the MinION DNA sequencer in-flight on the International Space Station (ISS), and benchmarked its performance off-Earth against the MinION, Illumina MiSeq, and PacBio RS II sequencing platforms in terrestrial laboratories. Samples contained equimolar mixtures of genomic DNA from lambda bacteriophage, Escherichia coli (strain K12, MG1655) and Mus musculus (female BALB/c mouse). Nine sequencing runs were performed aboard the ISS over a 6-month period, yielding a total of 276,882 reads with no apparent decrease in performance over time. From sequence data collected aboard the ISS, we constructed directed assemblies of the ~4.6 Mb E. coli genome, ~48.5 kb lambda genome, and a representative M. musculus sequence (the ~16.3 kb mitochondrial genome), at 100%, 100%, and 96.7% consensus pairwise identity, respectively; de novo assembly of the E. coli genome from raw reads yielded a single contig comprising 99.9% of the genome at 98.6% consensus pairwise identity. Simulated real-time analyses of in-flight sequence data using an automated bioinformatic pipeline and laptop-based genomic assembly demonstrated the feasibility of sequencing analysis and microbial identification aboard the ISS. These findings illustrate the potential for sequencing applications including disease diagnosis, environmental monitoring, and elucidating the molecular basis for how organisms respond to spaceflight.


Asunto(s)
Genoma , Nanoporos , Análisis de Secuencia de ADN/métodos , Vuelo Espacial , Animales , Escherichia coli/genética , Femenino , Genoma Bacteriano , Ratones , Ratones Endogámicos BALB C/genética
13.
PLoS One ; 10(4): e0125292, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25894343

RESUMEN

Vulvar Intraepithelial Neoplasia (VIN) is the precursor lesion of Vulvar Squamous Cell Carcinoma (VSCC), and the differentiated type (dVIN) is more frequently observed in relation to VSCC. In contrast to usual-type VIN (uVIN), which is related to infection by human papillomavirus (HPV), a germline mutation in the p53 gene is thought to be associated with ~90% of dVIN cases. To date, no infectious agent has been identified in association with dVIN, and studies investigating this possibility have been hindered by the difficulty in accurately diagnosing dVIN from small biopsies. Here, we used immunostaining for p16ink4a), a biomarker for HPV infection, to study 14 uVIN high-grade VIN and 14 dVIN cases, and to select 10 dVIN cases to broadly screen for all kn(own viruses using a pan-viral microarray platform (ViroChip). All of the uVIN tissue samples, including 8 warty and 6 basaloid cases, showed positivity with the p16(ink4a) immunostain. The staining pattern was full-thickness for all except two cases in which positive staining was localized in the lower 1/3 of the epidermis. In contrast, immunostaining for p16(ink4a) was negative in all dVIN cases. ViroChip analysis of 10 pure dVIN samples confirmed the absence of human papillomavirus subtypes or any other virus with the exception of a single sample that showed a weak microarray signature to a porcine herpesvirus. Follow-up PCR testing of the sample was negative for herpesvirus, and in-depth metagenomic next-generation sequencing revealed only sequences corresponding to non-pathogenic viral flora and bacterial contamination. In this study, we demonstrated lack of a virus association in 10 dVIN cases. Alternative pathways for carcinogenesis such as the p53 mutation should be considered for investigation of potential treatment options in dVIN.


Asunto(s)
Carcinoma in Situ/patología , Carcinoma in Situ/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Papillomaviridae/genética , Papillomaviridae/fisiología , Neoplasias de la Vulva/patología , Neoplasias de la Vulva/virología , Anciano , Carcinoma in Situ/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Persona de Mediana Edad , Papillomaviridae/aislamiento & purificación , Neoplasias de la Vulva/metabolismo
14.
Genome Med ; 7: 99, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26416663

RESUMEN

We report unbiased metagenomic detection of chikungunya virus (CHIKV), Ebola virus (EBOV), and hepatitis C virus (HCV) from four human blood samples by MinION nanopore sequencing coupled to a newly developed, web-based pipeline for real-time bioinformatics analysis on a computational server or laptop (MetaPORE). At titers ranging from 10(7)-10(8) copies per milliliter, reads to EBOV from two patients with acute hemorrhagic fever and CHIKV from an asymptomatic blood donor were detected within 4 to 10 min of data acquisition, while lower titer HCV virus (1 × 10(5) copies per milliliter) was detected within 40 min. Analysis of mapped nanopore reads alone, despite an average individual error rate of 24 % (range 8-49 %), permitted identification of the correct viral strain in all four isolates, and 90 % of the genome of CHIKV was recovered with 97-99 % accuracy. Using nanopore sequencing, metagenomic detection of viral pathogens directly from clinical samples was performed within an unprecedented <6 hr sample-to-answer turnaround time, and in a timeframe amenable to actionable clinical and public health diagnostics.


Asunto(s)
Virus Chikungunya/genética , Ebolavirus/genética , Hepacivirus/genética , ARN Viral/sangre , Análisis de Secuencia de ARN/métodos , Fiebre Chikungunya/sangre , Biología Computacional , Fiebre Hemorrágica Ebola/sangre , Hepatitis C/sangre , Humanos , Metagenómica , Nanoporos
15.
Lancet Infect Dis ; 15(6): 671-82, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25837569

RESUMEN

BACKGROUND: Enterovirus D68 was implicated in a widespread outbreak of severe respiratory illness across the USA in 2014 and has also been reported sporadically in patients with acute flaccid myelitis. We aimed to investigate the association between enterovirus D68 infection and acute flaccid myelitis during the 2014 enterovirus D68 respiratory outbreak in the USA. METHODS: Patients with acute flaccid myelitis who presented to two hospitals in Colorado and California, USA, between Nov 24, 2013, and Oct 11, 2014, were included in the study. Additional cases identified from Jan 1, 2012, to Oct 4, 2014, via statewide surveillance were provided by the California Department of Public Health. We investigated the cause of these cases by metagenomic next-generation sequencing, viral genome recovery, and enterovirus D68 phylogenetic analysis. We compared patients with acute flaccid myelitis who were positive for enterovirus D68 with those with acute flaccid myelitis but negative for enterovirus D68 using the two-tailed Fisher's exact test, two-sample unpaired t test, and Mann-Whitney U test. FINDINGS: 48 patients were included: 25 with acute flaccid myelitis, two with enterovirus-associated encephalitis, five with enterovirus-D68-associated upper respiratory illness, and 16 with aseptic meningitis or encephalitis who tested positive for enterovirus. Enterovirus D68 was detected in respiratory secretions from seven (64%) of 11 patients comprising two temporally and geographically linked acute flaccid myelitis clusters at the height of the 2014 outbreak, and from 12 (48%) of 25 patients with acute flaccid myelitis overall. Phylogenetic analysis revealed that all enterovirus D68 sequences associated with acute flaccid myelitis grouped into a clade B1 strain that emerged in 2010. Of six coding polymorphisms in the clade B1 enterovirus D68 polyprotein, five were present in neuropathogenic poliovirus or enterovirus D70, or both. One child with acute flaccid myelitis and a sibling with only upper respiratory illness were both infected by identical enterovirus D68 strains. Enterovirus D68 viraemia was identified in a child experiencing acute neurological progression of his paralytic illness. Deep metagenomic sequencing of cerebrospinal fluid from 14 patients with acute flaccid myelitis did not reveal evidence of an alternative infectious cause to enterovirus D68. INTERPRETATION: These findings strengthen the putative association between enterovirus D68 and acute flaccid myelitis and the contention that acute flaccid myelitis is a rare yet severe clinical manifestation of enterovirus D68 infection in susceptible hosts. FUNDING: National Institutes of Health, University of California, Abbott Laboratories, and the Centers for Disease Control and Prevention.


Asunto(s)
Brotes de Enfermedades , Infecciones por Enterovirus/complicaciones , Infecciones por Enterovirus/epidemiología , Enterovirus/aislamiento & purificación , Mielitis/complicaciones , Mielitis/epidemiología , Paraplejía/epidemiología , Adolescente , Adulto , Anciano , California/epidemiología , Niño , Preescolar , Estudios de Cohortes , Colorado/epidemiología , Biología Computacional , Enterovirus/clasificación , Enterovirus/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Metagenómica , Persona de Mediana Edad , Paraplejía/etiología , Filogenia , Estudios Retrospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA