Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Intervalo de año de publicación
1.
Antimicrob Resist Infect Control ; 11(1): 88, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35729671

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) causes community-acquired respiratory tract infections during winter. However, outbreaks in hospitals also occur repeatedly. In particular, patients with hematologic malignancies are at an increased risk for a severe and potentially fatal course of RSV infection. Here we present the investigation of an RSV outbreak in a hematology ward for adults following the ORION statement. METHODS: An epidemiologic and molecular outbreak analysis was performed. We developed and employed a minimal oligonucleotide probe set in target capture probe sequencing that allows cost-effective RSV-A or -B capturing to reconstruct RSV genomes from clinical samples. RESULTS: Four adult patients were involved in the outbreak caused by RSV-B in March 2019. The enforcement of the pre-existing infection control measures by effective training of hospital staff contributed to a successful containment. PCR-based RSV screening on the ward enabled early detection of new cases and rapid isolation measures. The molecular analysis demonstrated that the outbreak sequences were highly related and distinct to other RSV-B strains circulating at the same time. CONCLUSIONS: A multimodal infection control concept is essential for the timely detection and control of RSV outbreaks in patients with hematological disease. Among other measures, preventive screening for respiratory viruses is recommended. Furthermore, the integration of conventional and molecular epidemiology, such as whole-genome sequencing and variant calling, significantly contributes to the understanding of transmission pathways. Based on this, appropriate conclusions can be drawn for targeted prevention measures that have prepared us for the COVID-19 pandemic beyond the RSV approach described here.


Asunto(s)
COVID-19 , Infección Hospitalaria , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Adulto , Infección Hospitalaria/prevención & control , Brotes de Enfermedades , Humanos , Pandemias , Infecciones por Virus Sincitial Respiratorio/epidemiología , Virus Sincitial Respiratorio Humano/genética
2.
BMC Res Notes ; 9: 128, 2016 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-26921094

RESUMEN

Recessive mutations in the MPV17 gene cause mitochondrial DNA depletion syndrome, a fatal infantile genetic liver disease in humans. Loss of function in mice leads to glomerulosclerosis and sensineural deafness accompanied with mitochondrial DNA depletion. Mutations in the yeast homolog Sym1, and in the zebra fish homolog tra cause interesting, but not obviously related phenotypes, although the human gene can complement the yeast Sym1 mutation. The MPV17 protein is a hydrophobic membrane protein of 176 amino acids and unknown function. Initially localised in murine peroxisomes, it was later reported to be a mitochondrial inner membrane protein in humans and in yeast. To resolve this contradiction we tested two new mouse monoclonal antibodies directed against the human MPV17 protein in Western blots and immunohistochemistry on human U2OS cells. One of these monoclonal antibodies showed specific reactivity to a protein of 20 kD absent in MPV17 negative mouse cells. Immunofluorescence studies revealed colocalisation with peroxisomal, endosomal and lysosomal markers, but not with mitochondria. This data reveal a novel connection between a possible peroxisomal/endosomal/lysosomal function and mitochondrial DNA depletion.


Asunto(s)
Anticuerpos Monoclonales/química , Endosomas/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Peroxisomas/metabolismo , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Línea Celular Tumoral , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Endosomas/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Lisosomas/ultraestructura , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , Mutación , Osteoblastos/metabolismo , Osteoblastos/ultraestructura , Peroxisomas/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
J Virol Methods ; 203: 54-64, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24681050

RESUMEN

Hepatitis B virus (HBV) is hyperendemic to southern Africa, with genotype A of HBV being the predominant genotype, and subgenotype A1 prevailing. Infection with this subgenotype is associated with rapid disease progression, and high frequency of hepatocellular carcinoma development. The objectives of our study was to construct recombinant 1.28 mer replication competent HBV DNA plasmids of subgenotypes A1, A2 and D3 containing authentic endogenous HBV promoters and to follow their replication in vitro after transfection of Huh7 cells. We found that subgenotype D3 replicated at a lower level, as measured by HBsAg and HBV DNA levels, when compared to cells transfected with genotype A. There was no difference in the intracellular and extracellular HBsAg between cells transfected with subgenotypes A1 or A2. Cells transfected with subgenotype A1 had higher levels of intracellular replicative intermediates and HBcAg, and lower extracellular expression of HBeAg from days 1 to 3, when compared to cells transfected with subgenotype A2. In conclusion, the generation of these replication competent clones is an important step in the functional characterization of subgenotypes of HBV circulating in Africa and their comparison to strains circulating in other geographical regions of the world.


Asunto(s)
Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Regiones Promotoras Genéticas , Replicación Viral , Línea Celular , Genotipo , Antígenos de la Hepatitis B/análisis , Hepatocitos/virología , Humanos , Plásmidos , Transfección
4.
Nucleic Acids Res ; 40(22): 11363-79, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23034801

RESUMEN

Survival time-associated plant homeodomain (PHD) finger protein in Ovarian Cancer 1 (SPOC1, also known as PHF13) is known to modulate chromatin structure and is essential for testicular stem-cell differentiation. Here we show that SPOC1 is recruited to DNA double-strand breaks (DSBs) in an ATM-dependent manner. Moreover, SPOC1 localizes at endogenous repair foci, including OPT domains and accumulates at large DSB repair foci characteristic for delayed repair at heterochromatic sites. SPOC1 depletion enhances the kinetics of ionizing radiation-induced foci (IRIF) formation after γ-irradiation (γ-IR), non-homologous end-joining (NHEJ) repair activity, and cellular radioresistance, but impairs homologous recombination (HR) repair. Conversely, SPOC1 overexpression delays IRIF formation and γH2AX expansion, reduces NHEJ repair activity and enhances cellular radiosensitivity. SPOC1 mediates dose-dependent changes in chromatin association of DNA compaction factors KAP-1, HP1-α and H3K9 methyltransferases (KMT) GLP, G9A and SETDB1. In addition, SPOC1 interacts with KAP-1 and H3K9 KMTs, inhibits KAP-1 phosphorylation and enhances H3K9 trimethylation. These findings provide the first evidence for a function of SPOC1 in DNA damage response (DDR) and repair. SPOC1 acts as a modulator of repair kinetics and choice of pathways. This involves its dose-dependent effects on DNA damage sensors, repair mediators and key regulators of chromatin structure.


Asunto(s)
Cromatina/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Rayos gamma , Heterocromatina , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Tolerancia a Radiación , Reparación del ADN por Recombinación , Proteínas Represoras/metabolismo , Proteína 28 que Contiene Motivos Tripartito
5.
Proc Natl Acad Sci U S A ; 104(31): 12913-8, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17646655

RESUMEN

A surprisingly large population of mRNAs has been shown to localize to sensory axons, but few RNA-binding proteins have been detected in these axons. These axonal mRNAs include several potential binding targets for the La RNA chaperone protein. La is transported into axonal processes in both culture and peripheral nerve. Interestingly, La is posttranslationally modified in sensory neurons by sumoylation. In axons, small ubiquitin-like modifying polypeptides (SUMO)-La interacts with dynein, whereas native La interacts with kinesin. Lysine 41 is required for sumoylation, and sumoylation-incompetent La(K41R) shows only anterograde transport, whereas WT La shows both anterograde and retrograde transport in axons. Thus, sumoylation of La determines the directionality of its transport within the axonal compartment, with SUMO-La likely recycling to the cell body.


Asunto(s)
Transporte Axonal , Axones/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína SUMO-1/metabolismo , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Dineínas/metabolismo , Humanos , Cinesinas/metabolismo , Mucoproteínas/genética , Mucoproteínas/metabolismo , Mutación/genética , Unión Proteica , Transporte de Proteínas , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley , Nervio Ciático/metabolismo , Técnicas de Cultivo de Tejidos
6.
Nucleic Acids Res ; 34(1): 353-63, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16410615

RESUMEN

The posttranscriptional regulatory element (PRE) is considered to enhance hepatitis B virus (HBV) gene expression by facilitating the nuclear export of intronless viral subgenomic RNAs. Its role in the RNA metabolism of the viral pregenomic RNA (pgRNA) is currently unknown. We identified a positively cis-acting splicing regulatory element (SRE-1) and present two lines of evidence for its functionality. Firstly, in a heterologous context SRE-1 functionally substitutes for a retroviral bidirectional exonic splicing enhancer (ESE). As expected, SRE-1 is a splicing enhancer also in its natural viral sequence context, since deletion of SRE-1 reduces splicing of pgRNA in cell culture experiments. Secondly, we show that stimulation of HBV RNA splicing by the splicing factor PSF was repressed by the PRE. Analysis of a variety of PSF mutants indicated that RNA-binding and protein-protein interaction were required to enhance splicing. In addition, we show that the PRE contributed to pgRNA stability, but has little influence on its nuclear export. Herein, we report for the first time that the PRE harbors splicing stimulating and inhibiting regulatory elements controlling processing of the viral pregenome. We discuss a model in which the regulation of pgRNA splicing depends on cellular factors interacting with the PRE.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/genética , Empalme del ARN , ARN Viral/química , ARN Viral/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico , Línea Celular Tumoral , Virus de la Hepatitis B/metabolismo , Humanos , Factor de Empalme Asociado a PTB , Proteínas de Unión al ARN/metabolismo
7.
J Biol Chem ; 279(25): 26563-70, 2004 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-15060081

RESUMEN

Here we provide evidence for an interaction-dependent subnuclear trafficking of the human La (hLa) protein, known as transient interaction partner of a variety of RNAs. Among these, precursor transcripts of certain RNAs are located in the nucleoplasm or nucleolus. Here we examined which functional domains of hLa are involved in its nuclear trafficking. By using green fluorescent-hLa fusion proteins, we discovered a nucleolar localization signal and demonstrated its functionality in a heterologous context. In addition, we revealed that the RRM2 motif of hLa is essential both for its RNA binding competence in vitro and in vivo and its exit from the nucleolus. Our data imply that hLa traffics between different subnuclear compartments, which depend decisively on a functional nucleolar localization signal as well as on RNA binding. Directed trafficking of hLa is fully consistent with its function in the maturation of precursor RNAs located in different subnuclear compartments.


Asunto(s)
Transporte Activo de Núcleo Celular , Señales de Localización Nuclear , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Secuencia de Aminoácidos , Autoantígenos , Línea Celular Tumoral , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , ARN/química , ARN/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Transfección , Antígeno SS-B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA