Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Antimicrob Chemother ; 77(2): 474-482, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34741609

RESUMEN

BACKGROUND: Deep sequencing could improve understanding of HIV treatment failure and viral population dynamics. However, this tool is often inaccessible in low- and middle-income countries. OBJECTIVES: To determine the genetic patterns of resistance emerging in West African HIV-1 subtypes during first-line virological failure, and the implications for future antiretroviral options. PATIENTS AND METHODS: Participants were selected from a Nigerian cohort of people living with HIV who had failed first-line ART and subsequently switched to second-line therapy. Whole HIV-1 genome sequences were generated from first-line virological failure samples with Illumina MiSeq. Mutations detected at ≥2% frequency were analysed and compared by subtype. RESULTS: HIV-1 sequences were obtained from 101 participants (65% female, median age 30 years, median 32.9 months of nevirapine- or efavirenz-based ART). Thymidine analogue mutations (TAMs) were detected in 61%, other core NRTI mutations in 92% and NNRTI mutations in 99%. Minority variants (<20% frequency) comprised 18% of all mutations. K65R was more prevalent in CRF02_AG than G subtypes (33% versus 7%; P = 0.002), and ≥3 TAMs were more common in G than CRF02_AG (52% versus 24%; P = 0.004). Subtype G viruses also contained more RT cleavage site mutations. Cross-resistance to at least one of the newer NNRTIs, doravirine, etravirine or rilpivirine, was predicted in 81% of participants. CONCLUSIONS: Extensive drug resistance had accumulated in people with West African HIV-1 subtypes, prior to second-line ART. Deep sequencing significantly increased the detection of resistance-associated mutations. Caution should be used if considering newer-generation NNRTI agents in this setting.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , VIH-1 , Adulto , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Farmacorresistencia Viral/genética , Femenino , Genotipo , Infecciones por VIH/tratamiento farmacológico , VIH-1/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación , Nigeria , Insuficiencia del Tratamiento , Carga Viral
2.
Microbiol Spectr ; 9(3): e0061021, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34817282

RESUMEN

Phenotypic drug susceptibility testing (DST) for tuberculosis (TB) requires weeks to yield results. Although molecular tests rapidly detect drug resistance-associated mutations (DRMs), they are not scalable to cover the full genome and the many DRMs that can predict resistance. Whole-genome sequencing (WGS) methods are scalable, but if conducted directly on sputum, typically require a target enrichment step, such as nucleic acid amplification. We developed a targeted isothermal amplification-nanopore sequencing workflow for rapid prediction of drug resistance of TB isolates. We used recombinase polymerase amplification (RPA) to perform targeted isothermal amplification (37°C for 90 min) of three regions within the Mycobacterium tuberculosis genome, followed by nanopore sequencing on the MinION. We tested 29 mycobacterial genomic DNA extracts from patients with drug-resistant (DR) TB and compared our results to those of WGS by Illumina and phenotypic DST to evaluate the accuracy of prediction of resistance to rifampin and isoniazid. Amplification by RPA showed fidelity equivalent to that of high-fidelity PCR (100% concordance). Nanopore sequencing generated DRM predictions identical to those of WGS, with considerably faster sequencing run times of minutes rather than days. The sensitivity and specificity of rifampin resistance prediction for our workflow were 96.3% (95% confidence interval [CI], 81.0 to 99.9%) and 100.0% (95% CI, 15.8 to 100.0%), respectively. For isoniazid resistance prediction, the sensitivity and specificity were 100.0% (95% CI, 86.3 to 100.0%) and 100.0% (95% CI, 39.8 to 100.0%), respectively. The workflow consumable costs per sample are less than £100. Our rapid and low-cost drug resistance genotyping workflow provides accurate prediction of rifampin and isoniazid resistance, making it appropriate for use in resource-limited settings. IMPORTANCE Current methods for diagnosing drug-resistant tuberculosis are time consuming, resulting in delays in patients receiving treatment and in transmission onwards. They also require a high level of laboratory infrastructure, which is often only available at centralized facilities, resulting in further delays to diagnosis and additional barriers to deployment in resource-limited settings. This article describes a new workflow that can diagnose drug-resistant TB in a shorter time, with less equipment, and for a lower price than current methods. The amount of TB DNA is first increased without the need for bulky and costly thermocycling equipment. The DNA is then read using a portable sequencer called a MinION, which indicates whether there are tell-tale changes in the DNA that indicate whether the TB strain is drug resistant. Our workflow could play an important role in the future in the fight against the public health challenge that is TB drug resistance.


Asunto(s)
Antituberculosos/farmacología , Farmacorresistencia Bacteriana , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/genética , Secuenciación de Nanoporos/métodos , Tuberculosis Resistente a Múltiples Medicamentos/diagnóstico , Genotipo , Humanos , Isoniazida/farmacología , Mycobacterium tuberculosis/clasificación , Mycobacterium tuberculosis/aislamiento & purificación , Secuenciación de Nanoporos/economía , Reacción en Cadena de la Polimerasa , Rifampin/farmacología , Sensibilidad y Especificidad , Esputo/microbiología , Tuberculosis Resistente a Múltiples Medicamentos/microbiología , Flujo de Trabajo
3.
J Infect ; 83(6): 693-700, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34610391

RESUMEN

OBJECTIVES: Recently emerging SARS-CoV-2 variants have been associated with an increased rate of transmission within the community. We sought to determine whether this also resulted in increased transmission within hospitals. METHODS: We collected viral sequences and epidemiological data of patients with community and healthcare associated SARS-CoV-2 infections, sampled from 16th November 2020 to 10th January 2021, from nine hospitals participating in the COG-UK HOCI study. Outbreaks were identified using ward information, lineage and pairwise genetic differences between viral sequences. RESULTS: Mixed effects logistic regression analysis of 4184 sequences showed healthcare-acquired infections were no more likely to be identified as the Alpha variant than community acquired infections. Nosocomial outbreaks were investigated based on overlapping ward stay and SARS-CoV-2 genome sequence similarity. There was no significant difference in the number of patients involved in outbreaks caused by the Alpha variant compared to outbreaks caused by other lineages. CONCLUSIONS: We find no evidence to support it causing more nosocomial transmission than previous lineages. This suggests that the stringent infection prevention measures already in place in UK hospitals contained the spread of the Alpha variant as effectively as other less transmissible lineages, providing reassurance of their efficacy against emerging variants of concern.


Asunto(s)
COVID-19 , Infección Hospitalaria , Infección Hospitalaria/epidemiología , Hospitales , Humanos , SARS-CoV-2 , Reino Unido/epidemiología
4.
Lancet Infect Dis ; 21(9): 1246-1256, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33857406

RESUMEN

BACKGROUND: Emergence of variants with specific mutations in key epitopes in the spike protein of SARS-CoV-2 raises concerns pertinent to mass vaccination campaigns and use of monoclonal antibodies. We aimed to describe the emergence of the B.1.1.7 variant of concern (VOC), including virological characteristics and clinical severity in contemporaneous patients with and without the variant. METHODS: In this cohort study, samples positive for SARS-CoV-2 on PCR that were collected from Nov 9, 2020, for patients acutely admitted to one of two hospitals on or before Dec 20, 2020, in London, UK, were sequenced and analysed for the presence of VOC-defining mutations. We fitted Poisson regression models to investigate the association between B.1.1.7 infection and severe disease (defined as point 6 or higher on the WHO ordinal scale within 14 days of symptoms or positive test) and death within 28 days of a positive test and did supplementary genomic analyses in a cohort of chronically shedding patients and in a cohort of remdesivir-treated patients. Viral load was compared by proxy, using PCR cycle threshold values and sequencing read depths. FINDINGS: Of 496 patients with samples positive for SARS-CoV-2 on PCR and who met inclusion criteria, 341 had samples that could be sequenced. 198 (58%) of 341 had B.1.1.7 infection and 143 (42%) had non-B.1.1.7 infection. We found no evidence of an association between severe disease and death and lineage (B.1.1.7 vs non-B.1.1.7) in unadjusted analyses (prevalence ratio [PR] 0·97 [95% CI 0·72-1·31]), or in analyses adjusted for hospital, sex, age, comorbidities, and ethnicity (adjusted PR 1·02 [0·76-1·38]). We detected no B.1.1.7 VOC-defining mutations in 123 chronically shedding immunocompromised patients or in 32 remdesivir-treated patients. Viral load by proxy was higher in B.1.1.7 samples than in non-B.1.1.7 samples, as measured by cycle threshold value (mean 28·8 [SD 4·7] vs 32·0 [4·8]; p=0·0085) and genomic read depth (1280 [1004] vs 831 [682]; p=0·0011). INTERPRETATION: Emerging evidence exists of increased transmissibility of B.1.1.7, and we found increased virus load by proxy for B.1.1.7 in our data. We did not identify an association of the variant with severe disease in this hospitalised cohort. FUNDING: University College London Hospitals NHS Trust, University College London/University College London Hospitals NIHR Biomedical Research Centre, Engineering and Physical Sciences Research Council.


Asunto(s)
COVID-19/virología , Genoma Viral , SARS-CoV-2/genética , Índice de Severidad de la Enfermedad , Secuenciación Completa del Genoma , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Londres , Masculino , Persona de Mediana Edad , Filogenia , Reino Unido , Carga Viral , Esparcimiento de Virus
5.
Virus Evol ; 6(1): veaa004, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32395255

RESUMEN

Recombination is an important feature of HIV evolution, occurring both within and between the major branches of diversity (subtypes). The Ugandan epidemic is primarily composed of two subtypes, A1 and D, that have been co-circulating for 50 years, frequently recombining in dually infected patients. Here, we investigate the frequency of recombinants in this population and the location of breakpoints along the genome. As part of the PANGEA-HIV consortium, 1,472 consensus genome sequences over 5 kb have been obtained from 1,857 samples collected by the MRC/UVRI & LSHTM Research unit in Uganda, 465 (31.6 per cent) of which were near full-length sequences (>8 kb). Using the subtyping tool SCUEAL, we find that of the near full-length dataset, 233 (50.1 per cent) genomes contained only one subtype, 30.8 per cent A1 (n = 143), 17.6 per cent D (n = 82), and 1.7 per cent C (n = 8), while 49.9 per cent (n = 232) contained more than one subtype (including A1/D (n = 164), A1/C (n = 13), C/D (n = 9); A1/C/D (n = 13), and 33 complex types). K-means clustering of the recombinant A1/D genomes revealed a section of envelope (C2gp120-TMgp41) is often inherited intact, whilst a generalized linear model was used to demonstrate significantly fewer breakpoints in the gag-pol and envelope C2-TM regions compared with accessory gene regions. Despite similar recombination patterns in many recombinants, no clearly supported circulating recombinant form (CRF) was found, there was limited evidence of the transmission of breakpoints, and the vast majority (153/164; 93 per cent) of the A1/D recombinants appear to be unique recombinant forms. Thus, recombination is pervasive with clear biases in breakpoint location, but CRFs are not a significant feature, characteristic of a complex, and diverse epidemic.

6.
J Antimicrob Chemother ; 75(6): 1575-1579, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32105319

RESUMEN

OBJECTIVES: HIV-1 integrase inhibitors are recommended as first-line therapy by WHO, though efficacy and resistance data for non-B subtypes are limited. Two recent trials have identified the integrase L74I mutation to be associated with integrase inhibitor treatment failure in HIV-1 non-B subtypes. We sought to define the prevalence of integrase resistance mutations, including L74I, in West Africa. METHODS: We studied a Nigerian cohort of recipients prior to and during receipt of second-line PI-based therapy, who were integrase inhibitor-naive. Illumina next-generation sequencing with target enrichment was used on stored plasma samples. Drug resistance was interpreted using the Stanford Resistance Database and the IAS-USA 2019 mutation lists. RESULTS: Of 115 individuals, 59.1% harboured CRF02_AG HIV-1 and 40.9% harboured subtype G HIV-1. Four participants had major IAS-USA integrase resistance-associated mutations detected at low levels (2%-5% frequency). Two had Q148K minority variants and two had R263K (one of whom also had L74I). L74I was detected in plasma samples at over 2% frequency in 40% (46/115). Twelve (26.1%) had low-level minority variants of between 2% and 20% of the viral population sampled. The remaining 34 (73.9%) had L74I present at >20% frequency. L74I was more common among those with subtype G infection (55.3%, 26/47) than those with CRF02_AG infection (29.4%, 20/68) (P = 0.005). CONCLUSIONS: HIV-1 subtypes circulating in West Africa appear to have very low prevalence of major integrase mutations, but significant prevalence of L74I. A combination of in vitro and clinical studies is warranted to understand the potential implications.


Asunto(s)
Infecciones por VIH , Inhibidores de Integrasa VIH , Integrasa de VIH , VIH-1 , África Occidental , Farmacorresistencia Viral/genética , Genotipo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/epidemiología , Integrasa de VIH/genética , Inhibidores de Integrasa VIH/farmacología , Inhibidores de Integrasa VIH/uso terapéutico , VIH-1/genética , Humanos , Mutación , Prevalencia
7.
J Invest Dermatol ; 140(4): 774-784.e11, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31626786

RESUMEN

Varicella zoster virus (VZV) is a skin-tropic virus that infects epidermal keratinocytes and causes chickenpox. Although common, VZV infection can be life-threatening, particularly in the immunocompromized. Therefore, understanding VZV-keratinocyte interactions is important to find new treatments beyond vaccination and antiviral drugs. In VZV-infected skin, kallikrein 6 and the ubiquitin ligase MDM2 are upregulated concomitant with keratin 10 (KRT10) downregulation. MDM2 binds to KRT10, targeting it for degradation via the ubiquitin-proteasome pathway. Preventing KRT10 degradation reduced VZV propagation in culture and prevented epidermal disruption in skin explants. KRT10 knockdown induced expression of NR4A1 and enhanced viral propagation in culture. NR4A1 knockdown prevented viral propagation in culture, reduced LC3 levels, and increased LAMP2 expression. We therefore describe a drug-able pathway whereby MDM2 ubiquitinates and degrades KRT10, increasing NR4A1 expression and allowing VZV replication and propagation.


Asunto(s)
Regulación de la Expresión Génica , Herpes Zóster/genética , Herpes Zóster/metabolismo , Herpesvirus Humano 3/fisiología , Queratina-10/genética , Queratinocitos/patología , ARN/genética , Replicación Viral , Herpes Zóster/virología , Humanos , Queratina-10/biosíntesis , Queratinocitos/metabolismo , Queratinocitos/virología
8.
Influenza Other Respir Viruses ; 13(6): 556-563, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31536169

RESUMEN

BACKGROUND: The extent of transmission of influenza in hospital settings is poorly understood. Next generation sequencing may improve this by providing information on the genetic relatedness of viral strains. OBJECTIVES: We aimed to apply next generation sequencing to describe transmission in hospital and compare with methods based on routinely-collected data. METHODS: All influenza samples taken through routine care from patients at University College London Hospitals NHS Foundation Trust (September 2012 to March 2014) were included. We conducted Illumina sequencing and identified genetic clusters. We compared nosocomial transmission estimates defined using classical methods (based on time from admission to sample) and genetic clustering. We identified pairs of cases with space-time links and assessed genetic relatedness. RESULTS: We sequenced influenza sampled from 214 patients. There were 180 unique genetic strains, 16 (8.8%) of which seeded a new transmission chain. Nosocomial transmission was indicated for 32 (15.0%) cases using the classical definition and 34 (15.8%) based on genetic clustering. Of the 50 patients in a genetic cluster, 11 (22.0%) had known space-time links with other cases in the same cluster. Genetic distances between pairs of cases with space-time links were lower than for pairs without spatial links (P < .001). CONCLUSIONS: Genetic data confirmed that nosocomial transmission contributes significantly to the hospital burden of influenza and elucidated transmission chains. Prospective next generation sequencing could support outbreak investigations and monitor the impact of infection and control measures.


Asunto(s)
Infección Hospitalaria/transmisión , Infección Hospitalaria/virología , Gripe Humana/transmisión , Gripe Humana/virología , Orthomyxoviridae/fisiología , Adolescente , Adulto , Anciano , Niño , Preescolar , Análisis por Conglomerados , Infección Hospitalaria/epidemiología , Estudios Transversales , Femenino , Genoma Viral/genética , Hospitales , Humanos , Control de Infecciones , Gripe Humana/epidemiología , Londres/epidemiología , Masculino , Persona de Mediana Edad , Orthomyxoviridae/clasificación , Orthomyxoviridae/genética , ARN Viral/genética , Estudios Retrospectivos , Análisis de Secuencia de ADN , Adulto Joven
9.
J Infect Dis ; 218(9): 1485-1489, 2018 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-29873767

RESUMEN

Traditional epidemiological investigation of nosocomial transmission of influenza involves the identification of patients who have the same influenza virus type and who have overlapped in time and place. This method may misidentify transmission where it has not occurred or miss transmission when it has. We used influenza virus whole-genome sequencing (WGS) to investigate an outbreak of influenza A virus infection in a hematology/oncology ward and identified 2 separate introductions, one of which resulted in 5 additional infections and 79 bed-days lost. Results from WGS are becoming rapidly available and may supplement traditional infection control procedures in the investigation and management of nosocomial outbreaks.


Asunto(s)
Infección Hospitalaria/virología , Virus de la Influenza A/genética , Gripe Humana/virología , Infección Hospitalaria/epidemiología , Brotes de Enfermedades , Humanos , Control de Infecciones/métodos , Gripe Humana/epidemiología , Epidemiología Molecular/métodos , Secuenciación Completa del Genoma/métodos
10.
Cancer Cell ; 33(4): 620-633.e6, 2018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29634949

RESUMEN

The canine transmissible venereal tumor (CTVT) is a clonally transmissible cancer that regresses spontaneously or after treatment with vincristine, but we know little about the regression mechanisms. We performed global transcriptional, methylation, and functional pathway analyses on serial biopsies of vincristine-treated CTVTs and found that regression occurs in sequential steps; activation of the innate immune system and host epithelial tissue remodeling followed by immune infiltration of the tumor, arrest in the cell cycle, and repair of tissue damage. We identified CCL5 as a possible driver of CTVT regression. Changes in gene expression are associated with methylation changes at specific intragenic sites. Our results underscore the critical role of host innate immunity in triggering cancer regression.


Asunto(s)
Enfermedades de los Perros/tratamiento farmacológico , Perfilación de la Expresión Génica/veterinaria , Redes Reguladoras de Genes/efectos de los fármacos , Tumores Venéreos Veterinarios/tratamiento farmacológico , Vincristina/administración & dosificación , Animales , Puntos de Control del Ciclo Celular , Quimiocina CCL5/genética , Metilación de ADN , Enfermedades de los Perros/genética , Perros , Femenino , Regulación Neoplásica de la Expresión Génica , Inmunidad Innata/efectos de los fármacos , Masculino , Análisis de Secuencia de ARN/veterinaria , Tumores Venéreos Veterinarios/genética , Vincristina/farmacología
11.
PLoS One ; 13(2): e0192081, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29389981

RESUMEN

BACKGROUND & METHODS: The ICONIC project has developed an automated high-throughput pipeline to generate HIV nearly full-length genomes (NFLG, i.e. from gag to nef) from next-generation sequencing (NGS) data. The pipeline was applied to 420 HIV samples collected at University College London Hospitals NHS Trust and Barts Health NHS Trust (London) and sequenced using an Illumina MiSeq at the Wellcome Trust Sanger Institute (Cambridge). Consensus genomes were generated and subtyped using COMET, and unique recombinants were studied with jpHMM and SimPlot. Maximum-likelihood phylogenetic trees were constructed using RAxML to identify transmission networks using the Cluster Picker. RESULTS: The pipeline generated sequences of at least 1Kb of length (median = 7.46Kb, IQR = 4.01Kb) for 375 out of the 420 samples (89%), with 174 (46.4%) being NFLG. A total of 365 sequences (169 of them NFLG) corresponded to unique subjects and were included in the down-stream analyses. The most frequent HIV subtypes were B (n = 149, 40.8%) and C (n = 77, 21.1%) and the circulating recombinant form CRF02_AG (n = 32, 8.8%). We found 14 different CRFs (n = 66, 18.1%) and multiple URFs (n = 32, 8.8%) that involved recombination between 12 different subtypes/CRFs. The most frequent URFs were B/CRF01_AE (4 cases) and A1/D, B/C, and B/CRF02_AG (3 cases each). Most URFs (19/26, 73%) lacked breakpoints in the PR+RT pol region, rendering them undetectable if only that was sequenced. Twelve (37.5%) of the URFs could have emerged within the UK, whereas the rest were probably imported from sub-Saharan Africa, South East Asia and South America. For 2 URFs we found highly similar pol sequences circulating in the UK. We detected 31 phylogenetic clusters using the full dataset: 25 pairs (mostly subtypes B and C), 4 triplets and 2 quadruplets. Some of these were not consistent across different genes due to inter- and intra-subtype recombination. Clusters involved 70 sequences, 19.2% of the dataset. CONCLUSIONS: The initial analysis of genome sequences detected substantial hidden variability in the London HIV epidemic. Analysing full genome sequences, as opposed to only PR+RT, identified previously undetected recombinants. It provided a more reliable description of CRFs (that would be otherwise misclassified) and transmission clusters.


Asunto(s)
Genoma Viral , VIH-1/clasificación , Adulto , Femenino , VIH-1/genética , Humanos , Londres , Masculino , Persona de Mediana Edad , Filogenia , Recombinación Genética
12.
Proc Natl Acad Sci U S A ; 114(20): E3954-E3963, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28461481

RESUMEN

Most tissue-resident macrophages (Mφs) are believed to be derived prenatally and are assumed to maintain themselves throughout life by self-proliferation. However, in adult mice we identified a progenitor within bone marrow, early pro-B cell/fraction B, that differentiates into tissue Mφs. These Mφ precursors have non-rearranged B-cell receptor genes and coexpress myeloid (GR1, CD11b, and CD16/32) and lymphoid (B220 and CD19) lineage markers. During steady state, these precursors exit bone marrow, losing Gr1, and enter the systemic circulation, seeding the gastrointestinal system as well as pleural and peritoneal cavities but not the brain. While in these tissues, they acquire a transcriptome identical to embryonically derived tissue-resident Mφs. Similarly, these Mφ precursors also enter sites of inflammation, gaining CD115, F4/80, and CD16/32, and become indistinguishable from blood monocyte-derived Mφs. Thus, we have identified a population of cells within the bone marrow early pro-B cell compartment that possess functional plasticity to differentiate into either tissue-resident or inflammatory Mφs, depending on microenvironmental signals. We propose that these precursors represent an additional source of Mφ populations in adult mice during steady state and inflammation.


Asunto(s)
Activación de Macrófagos/fisiología , Macrófagos/inmunología , Células Precursoras de Linfocitos B/fisiología , Animales , Linfocitos B/fisiología , Médula Ósea , Células de la Médula Ósea/fisiología , Homeostasis/fisiología , Inflamación/inmunología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo
13.
Euro Surveill ; 22(8)2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28251889

RESUMEN

We report the molecular investigations of a large influenza A(H3N2) outbreak, in a season characterised by sharp increase in influenza admissions since December 2016. Analysis of haemagglutinin (HA) sequences demonstrated co-circulation of multiple clades (3C.3a, 3C.2a and 3C.2a1). Most variants fell into a novel subclade (proposed as 3C.2a2); they possessed four unique amino acid substitutions in the HA protein and loss of a potential glycosylation site. These changes potentially modify the H3N2 strain antigenicity.


Asunto(s)
Enfermedades Transmisibles Emergentes/genética , Epidemias , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Hemaglutininas/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Gripe Humana/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Sustitución de Aminoácidos , Niño , Preescolar , Enfermedades Transmisibles Emergentes/epidemiología , Femenino , Flujo Genético , Variación Genética , Glicosilación , Humanos , Lactante , Recién Nacido , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana/diagnóstico , Gripe Humana/virología , Londres/epidemiología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Filogenia , ARN Viral/genética , ARN Viral/aislamiento & purificación , Análisis de Secuencia de ADN , Adulto Joven
14.
J Infect Dis ; 212(1): 44-56, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25583169

RESUMEN

Early on in human immunodeficiency virus (HIV) type 1 infection, gut T-helper (Th) 17 cells are massively depleted leading eventually to compromised intestinal barrier function and excessive immune activation. In contrast, the functional Th17 cell compartment of the gut is well-maintained in nonpathogenic simian immunodeficiency virus infection as well as HIV-1 long-term nonprogressors. Here, we show that dendritic cells (DCs) loaded with HIV-1 bearing high surface complement levels after incubation in plasma from HIV-infected individuals secreted significantly higher concentrations of Th17-polarizing cytokines than DCs exposed to nonopsonized HIV-1. The enhanced Th17-polarizing capacity of in vitro-generated and BDCA-1(+) DCs directly isolated from blood was linked to activation of ERK. In addition, C3a produced from DCs exposed to complement-opsonized HIV was associated with the higher Th17 polarization. Our in vitro and ex vivo data, therefore, indicate that complement opsonization of HIV-1 strengthens DC-mediated antiviral immune functions by simultaneously triggering Th17 expansion and intrinsic C3 formation via DC activation.


Asunto(s)
Antígeno CD11b/análisis , Antígeno CD11c/análisis , Diferenciación Celular , Proteínas del Sistema Complemento/inmunología , Células Dendríticas/inmunología , VIH-1/inmunología , Células Th17/fisiología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad
15.
PLoS Pathog ; 10(12): e1004552, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25522326

RESUMEN

To date, no immunization of humans or animals has elicited broadly neutralizing sera able to prevent HIV-1 transmission; however, elicitation of broad and potent heavy chain only antibodies (HCAb) has previously been reported in llamas. In this study, the anti-HIV immune responses in immunized llamas were studied via deep sequencing analysis using broadly neutralizing monoclonal HCAbs as a guides. Distinct neutralizing antibody lineages were identified in each animal, including two defined by novel antibodies (as variable regions called VHH) identified by robotic screening of over 6000 clones. The combined application of five VHH against viruses from clades A, B, C and CRF_AG resulted in neutralization as potent as any of the VHH individually and a predicted 100% coverage with a median IC50 of 0.17 µg/ml for the panel of 60 viruses tested. Molecular analysis of the VHH repertoires of two sets of immunized animals showed that each neutralizing lineage was only observed following immunization, demonstrating that they were elicited de novo. Our results show that immunization can induce potent and broadly neutralizing antibodies in llamas with features similar to human antibodies and provide a framework to analyze the effectiveness of immunization protocols.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Sitios de Unión de Anticuerpos/inmunología , Antígenos CD4/inmunología , Camélidos del Nuevo Mundo/inmunología , Evolución Molecular , VIH-1/inmunología , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/uso terapéutico , Animales , Anticuerpos Neutralizantes/genética , Camélidos del Nuevo Mundo/genética , Modelos Animales de Enfermedad , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Mutación/genética
16.
PLoS One ; 9(10): e108384, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25290448

RESUMEN

Lymphoblastoid cell lines (LCLs) are commonly used in molecular genetics, supplying DNA for the HapMap and 1000 Genomes Projects, used to test chemotherapeutic agents, and informing the basis of a number of population genetics studies of gene expression. The process of transforming human B cells into LCLs requires the presence of Epstein-Barr virus (EBV), a double-stranded DNA virus which through B-cell immortalisation maintains an episomal virus genome in every cell of an LCL at variable copy numbers. Previous studies have reported that EBV alters host-gene expression and EBV copy number may be under host genetic control. We performed a genome-wide association study of EBV genome copy number in LCLs and found the phenotype to be highly heritable, although no individual SNPs achieved a significant association with EBV copy number. The expression of two host genes (CXCL16 and AGL) was positively correlated and expression of ADARB2 was negatively correlated with EBV copy number in a genotype-independent manner. This study shows an association between EBV copy number and the gene expression profile of LCLs, and suggests that EBV copy number should be considered as a covariate in future studies of host gene expression in LCLs.


Asunto(s)
Linfocitos B/metabolismo , Infecciones por Virus de Epstein-Barr/genética , Dosificación de Gen , Regulación de la Expresión Génica , Variación Genética , Herpesvirus Humano 4/genética , Linfocitos B/patología , Linfocitos B/virología , Línea Celular Transformada , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Estudio de Asociación del Genoma Completo , Herpesvirus Humano 4/inmunología , Humanos , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Transcriptoma
17.
PLoS Pathog ; 10(1): e1003896, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24497829

RESUMEN

Varicella zoster virus (VZV) is the etiological agent of chickenpox and shingles, diseases characterized by epidermal skin blistering. Using a calcium-induced keratinocyte differentiation model we investigated the interaction between epidermal differentiation and VZV infection. RNA-seq analysis showed that VZV infection has a profound effect on differentiating keratinocytes, altering the normal process of epidermal gene expression to generate a signature that resembles patterns of gene expression seen in both heritable and acquired skin-blistering disorders. Further investigation by real-time PCR, protein analysis and electron microscopy revealed that VZV specifically reduced expression of specific suprabasal cytokeratins and desmosomal proteins, leading to disruption of epidermal structure and function. These changes were accompanied by an upregulation of kallikreins and serine proteases. Taken together VZV infection promotes blistering and desquamation of the epidermis, both of which are necessary to the viral spread and pathogenesis. At the same time, analysis of the viral transcriptome provided evidence that VZV gene expression was significantly increased following calcium treatment of keratinocytes. Using reporter viruses and immunohistochemistry we confirmed that VZV gene and protein expression in skin is linked with cellular differentiation. These studies highlight the intimate host-pathogen interaction following VZV infection of skin and provide insight into the mechanisms by which VZV remodels the epidermal environment to promote its own replication and spread.


Asunto(s)
Diferenciación Celular , Varicela/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 3/fisiología , Queratinocitos/metabolismo , ARN Viral/biosíntesis , Proteínas Virales/biosíntesis , Replicación Viral/fisiología , Varicela/genética , Femenino , Humanos , Queratinocitos/patología , Queratinocitos/virología , Masculino , ARN Viral/genética , Análisis de Secuencia de ARN , Proteínas Virales/genética
18.
Mol Immunol ; 46(13): 2699-705, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19560824

RESUMEN

T cell activation is the final step in a complex pathway through which pathogen-derived peptide fragments can elicit an immune response. For it to occur, peptides must form stable complexes with Major Histocompatibility Complex (MHC) molecules and be presented on the cell surface. Computational predictors of MHC binding are often used within in silico vaccine design pathways. We have previously shown that, paradoxically, most bacterial proteins known experimentally to elicit an immune response in disease models are depleted in peptides predicted to bind to human MHC alleles. The results presented here, derived using software proven through benchmarking to be the most accurate currently available, show that vaccine antigens contain fewer predicted MHC-binding peptides than control bacterial proteins from almost all subcellular locations with the exception of cell wall and some cytoplasmic proteins. This effect is too large to be explained from the undoubted lack of precision of the software or from the amino acid composition of the antigens. Instead, we propose that pathogens have evolved under the influence of the host immune system so that surface proteins are depleted in potential MHC-binding peptides, and suggest that identification of a protein likely to contain a single immuno-dominant epitope is likely to be a productive strategy for vaccine design.


Asunto(s)
Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Programas Informáticos , Algoritmos , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Biología Computacional/métodos , Diseño de Fármacos , Epítopos de Linfocito T/química , Epítopos de Linfocito T/metabolismo , Antígenos HLA/inmunología , Antígenos HLA/metabolismo , Humanos , Unión Proteica , Reproducibilidad de los Resultados , Vacunas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA