Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transplant Proc ; 55(3): 623-628, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37024309

RESUMEN

PURPOSE: This study aims to assess the efficacy of current measurement strategies for lung sizing and the feasibility of future use of computed tomography (CT)-derived lung volumes to predict a donor-recipient lung size match during bilateral lung transplants. METHODS: We reviewed the data of 62 patients who underwent bilateral lung transplantation for interstitial lung disease and/or idiopathic pulmonary fibrosis from 2018 to 2019. Data for recipients was retrieved from the department's transplant database and medical records, and the donor's data was retrieved from the DonorNet. The data included demographic data, lung heights, measured total lung capacity (TLC) from plethysmography for recipients and estimated TLC for donors, clinical data, and CT-derived lung volumes in both pre- and post-transplant recipients. The post-transplant CT-derived lung volume in recipients was used as a surrogate for donor lung CT volumes due to inadequate or poor donor CT data. Computed tomography-derived lung volumes were calculated using thresholding, region growing, and cutting techniques on Computer-Aided Design and Mimics (Materialise NV, Leuven, Belgium) programs. Preoperative CT-derived lung volumes in recipients were compared with the plethysmography TLC, Frustum Model, and donor-predicted TLC. The ratio of the recipient's pre-and postoperative CT-derived volumes, the ratio of preoperative CT-derived lung volume, and donor-estimated TLC were studied to detect a correlation with 1-year outcomes. RESULTS: The recipient preoperative CT-derived volume correlated with the recipient preoperative plethysmography TLC (Pearson correlation coefficient [PCC] of 0.688) and with the recipient Frustum model volume (PCC of 0.593). The recipient postoperative CT-derived volume correlated with the recipient's postoperative plethysmography TLC (PCC of 0.651). There was no statistically significant correlation between recipients' CT-derived pre- or postoperative volume with donor-estimated TLC. The ratio of preoperative CT-derived volume to donor-estimated TLC correlated inversely with the length of ventilation (P value = .0031). The ratio of postoperative CT-derived volume to preoperative CT-derived volume correlated inversely with delayed sternal closure (P = .0039). No statistically significant correlations were found in evaluating outcomes related to lung oversizing in the recipient (defined as a postoperative to preoperative CT-derived lung volume ratio of >1.2). CONCLUSIONS: Generating CT-derived lung volumes is a valid and convenient method for evaluating lung volumes for transplantation in patients with ILD and/or IPF. Donor-estimated TLC should be interpreted carefully. Further studies should derive donor lung volumes from CT scans for a more accurate evaluation of lung size matching.


Asunto(s)
Fibrosis Pulmonar Idiopática , Enfermedades Pulmonares Intersticiales , Trasplante de Pulmón , Humanos , Mediciones del Volumen Pulmonar , Pulmón/diagnóstico por imagen , Pulmón/cirugía , Enfermedades Pulmonares Intersticiales/diagnóstico por imagen , Enfermedades Pulmonares Intersticiales/cirugía , Tomografía Computarizada por Rayos X , Fibrosis Pulmonar Idiopática/diagnóstico por imagen , Fibrosis Pulmonar Idiopática/cirugía
2.
J Virol ; 90(20): 9446-56, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27512068

RESUMEN

UNLABELLED: We have previously shown that IFIT1 is primarily responsible for the antiviral action of interferon (IFN) alpha/beta against parainfluenza virus type 5 (PIV5), selectively inhibiting the translation of PIV5 mRNAs. Here we report that while PIV2, PIV5, and mumps virus (MuV) are sensitive to IFIT1, nonrubulavirus members of the paramyxoviridae such as PIV3, Sendai virus (SeV), and canine distemper virus (CDV) are resistant. The IFIT1 sensitivity of PIV5 was not rescued by coinfection with an IFIT1-resistant virus (PIV3), demonstrating that PIV3 does not specifically inhibit the antiviral activity of IFIT1 and that the inhibition of PIV5 mRNAs is regulated by cis-acting elements. We developed an in vitro translation system using purified human IFIT1 to further investigate the mechanism of action of IFIT1. While the translations of PIV2, PIV5, and MuV mRNAs were directly inhibited by IFIT1, the translations of PIV3, SeV, and CDV mRNAs were not. Using purified human mRNA-capping enzymes, we show biochemically that efficient inhibition by IFIT1 is dependent upon a 5' guanosine nucleoside cap (which need not be N7 methylated) and that this sensitivity is partly abrogated by 2'O methylation of the cap 1 ribose. Intriguingly, PIV5 M mRNA, in contrast to NP mRNA, remained sensitive to inhibition by IFIT1 following in vitro 2'O methylation, suggesting that other structural features of mRNAs may influence their sensitivity to IFIT1. Thus, surprisingly, the viral polymerases (which have 2'-O-methyltransferase activity) of rubulaviruses do not protect these viruses from inhibition by IFIT1. Possible biological consequences of this are discussed. IMPORTANCE: Paramyxoviruses cause a wide variety of diseases, and yet most of their genes encode structural proteins and proteins involved in their replication cycle. Thus, the amount of genetic information that determines the type of disease that paramyxoviruses cause is relatively small. One factor that will influence disease outcomes is how they interact with innate host cell defenses, including the interferon (IFN) system. Here we show that different paramyxoviruses interact in distinct ways with cells in a preexisting IFN-induced antiviral state. Strikingly, all the rubulaviruses tested were sensitive to the antiviral action of ISG56/IFIT1, while all the other paramyxoviruses tested were resistant. We developed novel in vitro biochemical assays to investigate the mechanism of action of IFIT1, demonstrating that the mRNAs of rubulaviruses can be directly inhibited by IFIT1 and that this is at least partially because their mRNAs are not correctly methylated.


Asunto(s)
Proteínas Portadoras/farmacología , Paramyxoviridae/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Rubulavirus/genética , Células A549 , Proteínas Adaptadoras Transductoras de Señales , Línea Celular Tumoral , Humanos , Interferón-alfa/metabolismo , Metilación , Virus de la Parotiditis/genética , Virus de la Parainfluenza 5/genética , Caperuzas de ARN/genética , ARN Viral/genética , Proteínas de Unión al ARN , Virus Sendai/genética , Replicación Viral/genética
3.
J Virol ; 87(9): 4798-807, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23449801

RESUMEN

Preparations of parainfluenza virus 5 (PIV5) that are potent activators of the interferon (IFN) induction cascade were generated by high-multiplicity passage in order to accumulate defective interfering virus genomes (DIs). Nucleocapsid RNA from these virus preparations was extracted and subjected to deep sequencing. Sequencing data were analyzed using methods designed to detect internal deletion and "copyback" DIs in order to identify and characterize the different DIs present and to approximately quantify the ratio of defective to nondefective genomes. Trailer copybacks dominated the DI populations in IFN-inducing preparations of both the PIV5 wild type (wt) and PIV5-VΔC (a recombinant virus that does not encode a functional V protein). Although the PIV5 V protein is an efficient inhibitor of the IFN induction cascade, we show that nondefective PIV5 wt is unable to prevent activation of the IFN response by coinfecting copyback DIs due to the interfering effects of copyback DIs on nondefective virus protein expression. As a result, copyback DIs are able to very rapidly activate the IFN induction cascade prior to the expression of detectable levels of V protein by coinfecting nondefective virus.


Asunto(s)
Virus Defectuosos/genética , Genoma Viral , Infecciones por Rubulavirus/inmunología , Infecciones por Rubulavirus/virología , Rubulavirus/genética , Animales , Línea Celular , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Interferones/genética , Interferones/inmunología , Infecciones por Rubulavirus/genética , Proteínas Virales/genética
4.
J Gen Virol ; 93(Pt 2): 299-307, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22049094

RESUMEN

Conflicting reports exist regarding the requirement for virus replication in interferon (IFN) induction by paramyxoviruses. Our previous work has demonstrated that pathogen-associated molecular patterns capable of activating the IFN-induction cascade are not normally generated during virus replication, but are associated instead with the presence of defective interfering (DI) viruses. We demonstrate here that DIs of paramyxoviruses, including parainfluenza virus 5, mumps virus and Sendai virus, can activate the IFN-induction cascade and the IFN-ß promoter in the absence of virus protein synthesis. As virus protein synthesis is an absolute requirement for paramyxovirus genome replication, our results indicate that these DI viruses do not require replication to activate the IFN-induction cascade.


Asunto(s)
Interferón beta/biosíntesis , Interferón beta/genética , Paramyxoviridae/inmunología , Paramyxoviridae/fisiología , Regiones Promotoras Genéticas , Activación Transcripcional , Replicación Viral , Animales , Línea Celular , Virus Defectuosos/genética , Virus Defectuosos/inmunología , Humanos , Paramyxoviridae/genética , Rubulavirus , Proteínas Virales/biosíntesis
5.
Virology ; 415(1): 39-46, 2011 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-21511322

RESUMEN

It is generally thought that pathogen-associated molecular patterns (PAMPs) responsible for triggering interferon (IFN) induction are produced during virus replication and, to limit the activation of the IFN response by these PAMPs, viruses encode antagonists of IFN induction. Here we have studied the induction of IFN by parainfluenza virus type 5 (PIV5) at the single-cell level, using a cell line expressing GFP under the control of the IFN-ß promoter. We demonstrate that a recombinant PIV5 (termed PIV5-VΔC) that lacks a functional V protein (the viral IFN antagonist) does not activate the IFN-ß promoter in the majority of infected cells. We conclude that viral PAMPs capable of activating the IFN induction cascade are not produced or exposed during the normal replication cycle of PIV5, and suggest instead that defective viruses are primarily responsible for inducing IFN during PIV5 infection in this system.


Asunto(s)
Interferón beta/antagonistas & inhibidores , Interferón beta/genética , Regiones Promotoras Genéticas , Rubulavirus/fisiología , Proteínas Virales/fisiología , Animales , Línea Celular , Chlorocebus aethiops , Virus Defectuosos/genética , Virus Defectuosos/fisiología , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Immunoblotting , Interferón beta/metabolismo , Mutación , Rubulavirus/genética , Células Vero , Proteínas Virales/genética , Replicación Viral
6.
Virology ; 407(2): 247-55, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20833406

RESUMEN

The infection of cells by RNA viruses is associated with the recognition of virus PAMPs (pathogen-associated molecular patterns) and the production of type I interferon (IFN). To counter this, most, if not all, RNA viruses encode antagonists of the IFN system. Here we present data on the dynamics of IFN production and response during developing infections by paramyxoviruses, influenza A virus and bunyamwera virus. We show that only a limited number of infected cells are responsible for the production of IFN, and that this heterocellular production is a feature of the infecting virus as opposed to an intrinsic property of the cells.


Asunto(s)
Virus Bunyamwera/patogenicidad , Virus de la Influenza A/patogenicidad , Interferón Tipo I/metabolismo , Riñón/virología , Pulmón/virología , Paramyxoviridae/patogenicidad , Animales , Virus Bunyamwera/inmunología , Línea Celular Tumoral/virología , Chlorocebus aethiops , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/inmunología , Interferón Tipo I/genética , Interferón-alfa/genética , Interferón-alfa/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Riñón/citología , Riñón/inmunología , Pulmón/citología , Pulmón/inmunología , Paramyxoviridae/inmunología , Especificidad de la Especie , Células Vero/virología , Replicación Viral
7.
J Gen Virol ; 90(Pt 11): 2731-2738, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19625458

RESUMEN

Although the Enders strain of mumps virus (MuV) encodes a functional V protein that acts as an interferon (IFN) antagonist, in multi-cycle growth assays MuV Enders grew poorly in naïve ('IFN-competent' Hep2) cells but grew to high titres in 'IFN-compromised' Hep2 cells. Even so, the growth rate of MuV Enders was significantly slower in 'IFN-compromised' Hep2 cells when compared with its replication rate in Vero cells and with the replication rate of parainfluenza virus type 5 (a closely related paramyxovirus) in both naïve and 'IFN-compromised' Hep2 cells. This suggests that a consequence of slower growth is that the IFN system of naïve Hep2 cells can respond quickly enough to control the growth of MuV Enders. This is supported by the finding that rapidly growing variants of MuV Enders that were selected on 'IFN-compromised' Hep2 cells (i.e. in the absence of any selection pressure exerted by the IFN response) also grew to high titres on naïve Hep2 cells. Sequencing of the complete genome of one of these variants identified a single point mutation that resulted in a substitution of a conserved asparagine by histidine at position 498 of the haemagglutinin-neuraminidase protein, although this mutation was not present in all rapidly growing variants. These results support the concept that there is a race between the ability of a cell to detect and respond to virus infection and the ability of a virus to block the IFN response. Importantly, this emphasizes that factors other than viral IFN antagonists influence the sensitivity of viruses to IFN.


Asunto(s)
Interferones/antagonistas & inhibidores , Interferones/inmunología , Virus de la Parotiditis/inmunología , Virus de la Parotiditis/fisiología , Replicación Viral , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Chlorocebus aethiops , Análisis Mutacional de ADN , Proteína HN/genética , Humanos , Mutación Missense , Ensayo de Placa Viral
8.
J Gen Virol ; 90(Pt 9): 2147-56, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19458173

RESUMEN

Although the replication cycle of parainfluenza virus type 5 (PIV5) is initially severely impaired in cells in an interferon (IFN)-induced antiviral state, the virus still targets STAT1 for degradation. As a consequence, the cells can no longer respond to IFN and after 24-48 h, they go out of the antiviral state and normal virus replication is established. Following infection of cells in an IFN-induced antiviral state, viral nucleocapsid proteins are initially localized within small cytoplasmic bodies, and appearance of these cytoplasmic bodies correlates with the loss of STAT1 from infected cells. In situ hybridization, using probes specific for the NP and L genes, demonstrated the presence of virus genomes within these cytoplasmic bodies. These viral cytoplasmic bodies do not co-localize with cellular markers for stress granules, cytoplasmic P-bodies or autophagosomes. Furthermore, they are not large insoluble aggregates of viral proteins and/or nucleocapsids, as they can simply and easily be dispersed by 'cold-shocking' live cells, a process that disrupts the cytoskeleton. Given that during in vivo infections, PIV5 will inevitably infect cells in an IFN-induced antiviral state, we suggest that these cytoplasmic bodies are areas in which PIV5 genomes reside whilst the virus dismantles the antiviral state of the cells. Consequently, viral cytoplasmic bodies may play an important part in the strategy that PIV5 uses to circumvent the IFN system.


Asunto(s)
Citoplasma/inmunología , Genoma Viral , Cuerpos de Inclusión Viral/inmunología , Interferones/inmunología , Virus de la Parainfluenza 5/genética , Infecciones por Rubulavirus/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Citoplasma/genética , Citoplasma/virología , Humanos , Cuerpos de Inclusión Viral/genética , Interferones/genética , Virus de la Parainfluenza 5/inmunología , Virus de la Parainfluenza 5/fisiología , Infecciones por Rubulavirus/genética , Infecciones por Rubulavirus/virología , Células Vero , Replicación Viral
9.
Virology ; 365(1): 238-40, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17509637

RESUMEN

Whilst screening various cell lines for their ability to respond to interferon (IFN), we noted that in comparison to other tissue culture cells AGS tumour cells, which are widely used in biomedical research, had very low levels of STAT1. Subsequent analysis showed that the reason for this is that AGS cells are persistently infected with parainfluenza virus type 5 (PIV5; formally known as SV5), a virus that blocks the interferon (IFN) response by targeting STAT1 for proteasome-mediated degradation. Virus protein expression in AGS is altered in comparison to the normal pattern of virus protein synthesis observed in acutely infected cells, suggesting that the AGS virus is defective. We discuss the relevance of these results in terms of the need to screen cell lines for persistent virus infections that can alter cellular functions.


Asunto(s)
Células Cultivadas/virología , Paramyxoviridae/fisiología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Proteínas Estructurales Virales/metabolismo , Línea Celular , Interferones/metabolismo , Proteínas Estructurales Virales/genética
10.
J Gen Virol ; 86(Pt 1): 151-158, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15604442

RESUMEN

Previous work has documented that the V protein of simian virus 5 (SV5) targets STAT1 for proteasome-mediated degradation, whilst the V protein of human parainfluenza virus type 2 (hPIV2) targets STAT2. Here, it was shown that the processes of ubiquitination and degradation could be reconstructed in vitro by using programmed rabbit reticulocyte lysates. Using this system, the addition of bacterially expressed and purified SV5 V protein to programmed lysates was demonstrated to result in the polyubiquitination and degradation of in vitro-translated STAT1, but only if human STAT2 was also present. Surprisingly, in the same assay, purified hPIV2 V protein induced the polyubiquitination of both STAT1 and STAT2. In the light of these in vitro results, the specificity of degradation of STAT1 and STAT2 by SV5 and hPIV2 in tissue-culture cells was re-examined. As previously reported, STAT1 could not be detected in human cells that expressed SV5 V protein constitutively, whilst STAT2 could not be detected in human cells that expressed hPIV2 V protein, although the levels of STAT1 may also have been reduced in some human cells infected with hPIV2. In contrast, STAT1 could not be detected, whereas STAT2 remained present, in a variety of animal cells, including canine (MDCK) cells, that expressed the V protein of either SV5 or hPIV2. Thus, the V protein of SV5 appears to be highly specific for STAT1 degradation, but the V protein of hPIV2 is more promiscuous.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Respirovirus/metabolismo , Transactivadores/metabolismo , Proteínas Virales/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Línea Celular , Humanos , Conejos , Reticulocitos/metabolismo , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transducción de Señal , Especificidad de la Especie , Ubiquitina/metabolismo
11.
Proc Natl Acad Sci U S A ; 101(49): 17264-9, 2004 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-15563593

RESUMEN

Most paramyxoviruses circumvent the IFN response by blocking IFN signaling and limiting the production of IFN by virus-infected cells. Here we report that the highly conserved cysteine-rich C-terminal domain of the V proteins of a wide variety of paramyxoviruses binds melanoma differentiation-associated gene 5 (mda-5) product. mda-5 is an IFN-inducible host cell DExD/H box helicase that contains a caspase recruitment domain at its N terminus. Overexpression of mda-5 stimulated the basal activity of the IFN-beta promoter in reporter gene assays and significantly enhanced the activation of the IFN-beta promoter by intracellular dsRNA. Both these activities were repressed by coexpression of the V proteins of simian virus 5, human parainfluenza virus 2, mumps virus, Sendai virus, and Hendra virus. Similar results to the reporter assays were obtained by measuring IFN production. Inhibition of mda-5 by RNA interference or by dominant interfering forms of mda-5 significantly inhibited the activation of the IFN-beta promoter by dsRNA. It thus appears that mda-5 plays a central role in an intracellular signal transduction pathway that can lead to the activation of the IFN-beta promoter, and that the V proteins of paramyxoviruses interact with mda-5 to block its activity.


Asunto(s)
Interferón beta/genética , Paramyxoviridae/química , ARN Helicasas/antagonistas & inhibidores , Activación Transcripcional/efectos de los fármacos , Proteínas Virales/metabolismo , Animales , Sitios de Unión , Línea Celular , ARN Helicasas DEAD-box , Humanos , Helicasa Inducida por Interferón IFIH1 , Regiones Promotoras Genéticas/efectos de los fármacos , Unión Proteica , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Interferente Pequeño/farmacología , Transducción de Señal , Transfección , Proteínas Virales/genética , Proteínas Virales/farmacología
12.
J Virol ; 77(3): 2174-81, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12525652

RESUMEN

The V protein of the paramyxovirus simian virus 5 blocks interferon (IFN) signaling by targeting STAT1 for proteasome-mediated degradation. Here we report on the isolation of human cell lines that express the V protein and can no longer respond to IFN. A variety of viruses, particularly slow-growing wild-type viruses and vaccine candidate viruses (which are attenuated due to mutations that affect virus replication, virus spread, or ability to circumvent the IFN response), form bigger plaques and grow to titers that are increased as much as 10- to 4,000-fold in these IFN-nonresponsive cells. We discuss the practical applications of using such cells in vaccine development and manufacture, virus diagnostics and isolation of newly emerging viruses, and studies on host cell tropism and pathogenesis.


Asunto(s)
Interferones/farmacología , Transfección , Replicación Viral , Virus/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Humanos , Vacunas Sintéticas/inmunología , Células Vero , Vacunas Virales/inmunología
13.
J Virol ; 76(22): 11379-86, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12388698

RESUMEN

The V protein of simian virus 5 (SV5) blocks interferon signaling by targeting STAT1 for proteasome-mediated degradation. Here we present three main pieces of evidence which demonstrate that the p127 subunit (DDB1) of the UV damage-specific DNA binding protein (DDB) plays a central role in this degradation process. First, the V protein of an SV5 mutant which fails to target STAT1 for degradation does not bind DDB1. Second, mutations in the N and C termini of V which abolish the binding of V to DDB1 also prevent V from blocking interferon (IFN) signaling. Third, treatment of HeLa/SV5-V cells, which constitutively express the V protein of SV5 and thus lack STAT1, with short interfering RNAs specific for DDB1 resulted in a reduction in DDB1 levels with a concomitant increase in STAT1 levels and a restoration of IFN signaling. Furthermore, STAT1 is degraded in GM02415 (2RO) cells, which have a mutation in DDB2 (the p48 subunit of DDB) which abolishes its ability to interact with DDB1, thereby demonstrating that the role of DDB1 in STAT1 degradation is independent of its association with DDB2. Evidence is also presented which demonstrates that STAT2 is required for the degradation of STAT1 by SV5. These results suggest that DDB1, STAT1, STAT2, and V may form part of a large multiprotein complex which leads to the targeted degradation of STAT1 by the proteasome.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Rubulavirus/patogenicidad , Transactivadores/metabolismo , Proteínas Virales , Proteínas Estructurales Virales/metabolismo , Animales , Línea Celular , Células HeLa , Humanos , Interferones/metabolismo , Virus de la Parainfluenza 2 Humana/patogenicidad , Virus de la Parainfluenza 2 Humana/fisiología , Rubulavirus/fisiología , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transducción de Señal , Replicación Viral , Xerodermia Pigmentosa/metabolismo
14.
J Virol ; 76(5): 2159-67, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11836393

RESUMEN

Human cell lines were isolated that express the V protein of either simian virus 5 (SV5) or human parainfluenza virus type 2 (hPIV2); the cell lines were termed 2f/SV5-V and 2f/PIV2-V, respectively. STAT1 was not detectable in 2f/SV5-V cells, and the cells failed to signal in response to either alpha/beta interferons (IFN-alpha and IFN-beta, or IFN-alpha/beta) or gamma interferon (IFN-gamma). In contrast, STAT2 was absent from 2f/PIV2-V cells, and IFN-alpha/beta but not IFN-gamma signaling was blocked in these cells. Treatment of both 2f/SV5-V and 2f/PIV2-V cells with a proteasome inhibitor allowed the respective STAT levels to accumulate at rates similar to those seen in 2fTGH cells, indicating that the V proteins target the STATs for proteasomal degradation. Infection with SV5 can lead to a complete loss of both phosphorylated and nonphosphorylated forms of STAT1 by 6 h postinfection. Since the turnover of STAT1 in uninfected cells is longer than 24 h, we conclude that degradation of STAT1 is the main mechanism by which SV5 blocks interferon (IFN) signaling. Pretreatment of 2fTGH cells with IFN-alpha severely inhibited both SV5 and hPIV2 protein synthesis. However, and in marked contrast, pretreatment of 2fTGH cells with IFN-gamma had little obvious effect on SV5 protein synthesis but did significantly reduce the replication of hPIV2. Pretreament with IFN-alpha or IFN-gamma did not induce an antiviral state in 2f/SV5-V cells, indicating either that the induction of an antiviral state is completely dependent on STAT signaling or that the V protein interferes with other, STAT-independent cell signaling pathways that may be induced by IFNs. Even though SV5 blocked IFN signaling, the addition of exogenous IFN-alpha to the culture medium of 2fTGH cells 12 h after a low-multiplicity infection with SV5 significantly reduced the subsequent cell-to-cell spread of virus. The significance of the results in terms of the strategy that these viruses have evolved to circumvent the IFN response is discussed.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Virus de la Parainfluenza 2 Humana/fisiología , Rubulavirus/fisiología , Transactivadores/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Antivirales/farmacología , División Celular , Línea Celular , Clonación Molecular , Humanos , Interferones/farmacología , Virus de la Parainfluenza 2 Humana/efectos de los fármacos , Rubulavirus/efectos de los fármacos , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transfección , Rayos Ultravioleta , Proteínas Virales/metabolismo , Proteínas Estructurales Virales/genética , Replicación Viral
15.
J Virol ; 75(7): 3363-70, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11238862

RESUMEN

Previous work has demonstrated that the V protein of simian virus 5 (SV5) targets STAT1 for proteasome-mediated degradation (thereby blocking interferon [IFN] signaling) in human but not in murine cells. In murine BF cells, SV5 establishes a low-grade persistent infection in which the virus fluxes between active and repressed states in response to local production of IFN. Upon passage of persistently infected BF cells, virus mutants were selected that were better able to replicate in murine cells than the parental W3 strain of SV5 (wild type [wt]). Viruses with mutations in the Pk region of the N-terminal domain of the V protein came to predominate the population of viruses carried in the persistently infected cell cultures. One of these mutant viruses, termed SV5 mci-2, was isolated. Sequence analysis of the V/P gene of SV5 mci-2 revealed two nucleotide differences compared to wt SV5, only one of which resulted in an amino acid substitution (asparagine [N], residue 100, to aspartic acid [D]) in V. Unlike the protein of wt SV5, the V protein of SV5 mci-2 blocked IFN signaling in murine cells. Since the SV5 mci-2 virus had additional mutations in genes other than the V/P gene, a recombinant virus (termed rSV5-V/P N(100)D) was constructed that contained this substitution alone within the wt SV5 backbone to evaluate what effect the asparagine-to-aspartic-acid substitution in V had on the virus phenotype. In contrast to wt SV5, rSV5-V/P N(100)D blocked IFN signaling in murine cells. Furthermore, rSV5-V/P N(100)D virus protein synthesis in BF cells continued for significantly longer periods than that for wt SV5. However, even in cells infected with rSV5-V/P N(100)D, there was a late, but significant, inhibition in virus protein synthesis. Nevertheless, there was an increase in virus yield from BF cells infected with rSV5-V/P N(100)D compared to wt SV5, demonstrating a clear selective advantage to SV5 in being able to block IFN signaling in these cells.


Asunto(s)
Interferones/farmacología , Respirovirus/fisiología , Proteínas Estructurales Virales/fisiología , Animales , Secuencia de Bases , Proteínas de Unión al ADN/análisis , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Mutación Puntual , Respirovirus/genética , Factor de Transcripción STAT1 , Relación Estructura-Actividad , Transactivadores/análisis , Proteínas Virales/biosíntesis , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/genética
16.
Virology ; 269(2): 383-90, 2000 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-10753717

RESUMEN

STAT1 and STAT2 are cellular transcription factors involved in interferon (IFN) signaling and are thus critical for the IFN-induced antiviral state. We have previously shown that the paramyxovirus Simian Virus 5 (SV5) blocks both type I and type II interferon (IFN) signaling by targeting STAT1 for proteasome-mediated degradation. To determine whether this is a feature common to all Paramyxoviridae, we examined the abilities of SV5, Sendai virus (SeV), human respiratory syncytial virus (RSV), and human parainfluenza viruses types 2 and 3 (hPIV2 and hPIV3, respectively) to block interferon signaling. The results showed that in reporter assays SV5, SeV, and hPIV3 blocked both type I and type II IFN-signaling; hPIV2 blocked type I but not type II IFN-signaling; and RSV failed to block either type I or type II IFN-signaling. In agreement with these results, SV5 and SeV inhibited the formation of the ISGF3 and GAF transcription complexes (essential for type I and type II signaling, respectively). Surprisingly, although hPIV3 inhibited IFN-induction of the ISGF3 complex, GAF complexes were detected in hPIV3-infected cells. hPIV2 also blocked the formation of the ISGF3 complex but not the GAF complex, whereas RSV failed to block the induction of either complex. SV5 was the only virus that caused the degradation of STAT1. Indeed, in SeV- and hPIV3-infected cells STAT1 was phosphorylated on tyrosine 701 (Y701), a characteristic of IFN receptor activation. However, consistent with these viruses blocking IFN signaling downstream of receptor activation, there was a specific reduction in the levels of serine 727 (S727)-phosphorylated forms of STAT1alpha in SeV- and hPIV3-infected cells. In contrast both (Y701)- and (S727)-phosphorylated forms of STAT1 were detected in hPIV2-infected cells but there was a specific loss of STAT2. Both STAT1 (including Y701- and S727-phosphorylated forms) and STAT2 could readily be detected in RSV-infected cells. Despite not being able to block type I or type II IFN signaling, RSV was able to replicate in human cells that produce and respond to IFN, suggesting that RSV must have an alternative method(s) for circumventing the IFN response. These results demonstrate that, although interference with IFN signaling is a common strategy among Paramyxovirinae, distinct virus-specific mechanisms are used to achieve this end.


Asunto(s)
Antivirales/uso terapéutico , Interferones/uso terapéutico , Paramyxoviridae/fisiología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Humanos , Virus de la Parainfluenza 2 Humana/fisiología , Virus de la Parainfluenza 3 Humana/fisiología , Regiones Promotoras Genéticas , Virus Sincitiales Respiratorios/fisiología , Respirovirus/fisiología , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transducción de Señal , Transactivadores/metabolismo , Replicación Viral
17.
Diabetes Technol Ther ; 2(4): 541-8, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11469617

RESUMEN

A vacuum-lancet device was applied to the forearm for the purpose of obtaining capillary blood samples for glucose monitoring with minimal pain. In four clinical trials, a total of 215 individuals aged 12-77 years were tested four times using standard conditions and four times with either a different depth of lancing, different brand of lancet or a larger-sized device. The volume of blood collected using one-half atmosphere of vacuum in 40 sec was measured. The sensation and visual appearance of each lancet puncture on the forearm was recorded. Glucose was measured in forearm and in conventional fingerstick blood samples. The distribution of volumes was skewed to higher values with median values for each trial in the range of 3-10 microL. Ninety-five percent of the lancet sticks were judged as less painful than a fingerstick. Redness and bruising around the lanced sites were noted in some patients but disappeared within a few days. Overall correlation of the forearm versus fingerstick glucose values was 0.96. The vacuum-lancet device was very successful in obtaining capillary blood samples for glucose testing in a relatively painless manner. Incorporation of a glucose measuring system into the device might improve testing compliance among those who fear pain or the sight of blood.


Asunto(s)
Glucemia/análisis , Recolección de Muestras de Sangre/efectos adversos , Recolección de Muestras de Sangre/instrumentación , Monitoreo Fisiológico/métodos , Adolescente , Adulto , Distribución por Edad , Factores de Edad , Anciano , Recolección de Muestras de Sangre/métodos , Niño , Diseño de Equipo , Dedos/irrigación sanguínea , Antebrazo/irrigación sanguínea , Humanos , Persona de Mediana Edad , Dolor
18.
J Virol ; 73(12): 9928-33, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10559305

RESUMEN

To replicate in vivo, viruses must circumvent cellular antiviral defense mechanisms, including those induced by the interferons (IFNs). Here we demonstrate that simian virus 5 (SV5) blocks IFN signalling in human cells by inhibiting the formation of the IFN-stimulated gene factor 3 and gamma-activated factor transcription complexes that are involved in activating IFN-alpha/beta- and IFN-gamma-responsive genes, respectively. SV5 inhibits the formation of these complexes by specifically targeting STAT1, a component common to both transcription complexes, for proteasome-mediated degradation. Expression of the SV5 structural protein V, in the absence of other virus proteins, also inhibited IFN signalling and induced the degradation of STAT1. Following infection with SV5, STAT1 was degraded in the absence of virus protein synthesis and remained undetectable for up to 4 days postinfection. Furthermore, STAT1 was also degraded in IFN-pretreated cells, even though the cells were in an antiviral state. Since pretreatment of cells with IFN delayed but did not prevent virus replication and protein synthesis, these observations suggest that following infection of IFN-pretreated cells, SV5 remains viable within the cells until they eventually go out of the antiviral state.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Interferón Tipo I/metabolismo , Interferón beta/metabolismo , Interferón gamma/metabolismo , Complejos Multienzimáticos/metabolismo , Respirovirus , Transducción de Señal , Transactivadores/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Línea Celular , Humanos , Interferón Tipo I/farmacología , Factor 3 de Genes Estimulados por el Interferón , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón , Interferón-alfa , Interferón gamma/farmacología , Complejo de la Endopetidasa Proteasomal , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes , Factor de Transcripción STAT1 , Factores de Transcripción/metabolismo
19.
J Virol ; 73(4): 3125-33, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10074164

RESUMEN

Sendai virus (SeV) is highly pathogenic for mice. In contrast, mice (including SCID mice) infected with simian virus 5 (SV5) showed no overt signs of disease. Evidence is presented that a major factor which prevented SV5 from productively infecting mice was its inability to circumvent the interferon (IFN) response in mice. Thus, in murine cells that produce and respond to IFN, SV5 protein synthesis was rapidly switched off. In marked contrast, once SeV protein synthesis began, it continued, even if the culture medium was supplemented with alpha/beta IFN (IFN-alpha/beta). However, in human cells, IFN-alpha/beta did not inhibit the replication of either SV5 or SeV once virus protein synthesis was established. To begin to address the molecular basis for these observations, the effects of SeV and SV5 infections on the activation of an IFN-alpha/beta-responsive promoter and on that of the IFN-beta promoter were examined in transient transfection experiments. The results demonstrated that (i) SeV, but not SV5, inhibited an IFN-alpha/beta-responsive promoter in murine cells; (ii) both SV5 and SeV inhibited the activation of an IFN-alpha/beta-responsive promoter in human cells; and (iii) in both human and murine cells, SeV was a strong inducer of the IFN-beta promoter, whereas SV5 was a poor inducer. The ability of SeV and SV5 to inhibit the activation of IFN-responsive genes in human cells was confirmed by RNase protection experiments. The importance of these results in terms of paramyxovirus pathogenesis is discussed.


Asunto(s)
Regulación Viral de la Expresión Génica , Interferón-alfa/genética , Interferón beta/genética , Infecciones por Respirovirus/genética , Respirovirus/fisiología , Animales , Línea Celular , Humanos , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Replicación Viral/genética
20.
Motor Control ; 2(2): 106-13, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9644281

RESUMEN

Seven right-handed participants performed bimanual circling movements in either a symmetrical or an asymmetrical coordination mode. Movements were paced with an auditory metronome at predetermined frequencies corresponding to transition frequency, where asymmetrical patterns became unstable, or at two-thirds transition frequency where both symmetrical and asymmetrical patterns were stable. The pacing tones were presented in either a high (1000 Hz) or low (500 Hz) pitch, and the percentage of high-pitched tones during a 20 s trial varied between 0% and 70%. Participants were instructed to count the number of high-pitched pacing tones that occurred during a trial of bimanual circling. Overall, the symmetrical pattern was more stable than the asymmetrical pattern at both frequencies. Errors on the tone-counting task were significantly higher during asymmetrical circling than symmetrical circling but only at the transition movement frequency. The results suggest that cognitive processes play a role in maintaining coordination patterns within regions of instability.


Asunto(s)
Atención , Destreza Motora/fisiología , Estimulación Acústica , Análisis de Varianza , Femenino , Lateralidad Funcional , Humanos , Movimiento/fisiología , Desempeño Psicomotor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...