Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 1041-1056, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29885363

RESUMEN

The hepatitis C virus (HCV) life cycle is tightly linked to the host cell lipid metabolism with the endoplasmic reticulum-derived membranous web harboring viral RNA replication complexes and lipid droplets as virion assembly sites. To investigate HCV-induced changes in the lipid composition, we performed quantitative shotgun lipidomic studies of whole cell extracts and subcellular compartments. Our results indicate that HCV infection reduces the ratio of neutral to membrane lipids. While the amount of neutral lipids and lipid droplet morphology were unchanged, membrane lipids, especially cholesterol and phospholipids, accumulated in the microsomal fraction in HCV-infected cells. In addition, HCV-infected cells had a higher relative abundance of phosphatidylcholines and triglycerides with longer fatty acyl chains and a strikingly increased utilization of C18 fatty acids, most prominently oleic acid (FA [18:1]). Accordingly, depletion of fatty acid elongases and desaturases impaired HCV replication. Moreover, the analysis of free fatty acids revealed increased levels of polyunsaturated fatty acids (PUFAs) caused by HCV infection. Interestingly, inhibition of the PUFA synthesis pathway via knockdown of the rate-limiting Δ6-desaturase enzyme or by treatment with a high dose of a small-molecule inhibitor impaired viral progeny production, indicating that elevated PUFAs are needed for virion morphogenesis. In contrast, pretreatment with low inhibitor concentrations promoted HCV translation and/or early RNA replication. Taken together our results demonstrate the complex remodeling of the host cell lipid metabolism induced by HCV to enhance both virus replication and progeny production.


Asunto(s)
Hepacivirus/metabolismo , Hepatocitos/metabolismo , Interacciones Huésped-Patógeno , Metabolismo de los Lípidos/genética , Metaboloma , Virión/metabolismo , Replicación Viral/fisiología , Acetiltransferasas/antagonistas & inhibidores , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Línea Celular Tumoral , Colesterol/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Ácido Graso Desaturasas/antagonistas & inhibidores , Ácido Graso Desaturasas/genética , Ácido Graso Desaturasas/metabolismo , Elongasas de Ácidos Grasos , Ácidos Grasos Insaturados/metabolismo , Regulación de la Expresión Génica , Hepacivirus/crecimiento & desarrollo , Hepatocitos/química , Hepatocitos/virología , Humanos , Gotas Lipídicas/metabolismo , Gotas Lipídicas/virología , Microsomas/metabolismo , Microsomas/virología , Ácido Oléico/metabolismo , Fosfatidilcolinas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Viral/biosíntesis , ARN Viral/genética , Triglicéridos/metabolismo , Virión/crecimiento & desarrollo , Ensamble de Virus/fisiología
2.
PLoS One ; 10(3): e0117963, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25734423

RESUMEN

RIG-I pathway signaling of innate immunity against RNA virus infection is organized between the ER and mitochondria on a subdomain of the ER called the mitochondrial-associated ER membrane (MAM). The RIG-I adaptor protein MAVS transmits downstream signaling of antiviral immunity, with signaling complexes assembling on the MAM in association with mitochondria and peroxisomes. To identify components that regulate MAVS signalosome assembly on the MAM, we characterized the proteome of MAM, ER, and cytosol from cells infected with either chronic (hepatitis C) or acute (Sendai) RNA virus infections, as well as mock-infected cells. Comparative analysis of protein trafficking dynamics during both chronic and acute viral infection reveals differential protein profiles in the MAM during RIG-I pathway activation. We identified proteins and biochemical pathways recruited into and out of the MAM in both chronic and acute RNA viral infections, representing proteins that drive immunity and/or regulate viral replication. In addition, by using this comparative proteomics approach, we identified 3 new MAVS-interacting proteins, RAB1B, VTN, and LONP1, and defined LONP1 as a positive regulator of the RIG-I pathway. Our proteomic analysis also reveals a dynamic cross-talk between subcellular compartments during both acute and chronic RNA virus infection, and demonstrates the importance of the MAM as a central platform that coordinates innate immune signaling to initiate immunity against RNA virus infection.


Asunto(s)
Retículo Endoplásmico/metabolismo , Hepacivirus/fisiología , Mitocondrias/metabolismo , Proteoma/análisis , Virus Sendai/fisiología , Espectrometría de Masas en Tándem , Línea Celular , Cromatografía Líquida de Alta Presión , Retículo Endoplásmico/virología , Humanos , Redes y Vías Metabólicas , Microsomas/metabolismo , Microsomas/virología , Mitocondrias/virología , Análisis de Componente Principal , Replicación Viral
3.
Virus Res ; 168(1-2): 48-55, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22727684

RESUMEN

The replication of tick-borne encephalitis virus (TBEV), like that of all flaviviruses, is absolutely dependent on proteolytic processing. Production of the mature proteins C and prM from their common precursor requires the activity of the viral NS2B/3 protease (NS2B/3(pro)) at the C-terminus of protein C and the host signal peptidase I (SPaseI) at the N-terminus of protein prM. Recently, we have shown in cell culture that the cleavage of protein C and the subsequent production of TBEV particles can be made dependent on the activity of the foot-and-mouth disease virus 3C protease, but not on the activity of the HIV-1 protease (HIV1(pro)) (Schrauf et al., 2012). To investigate this failure, we developed an in vitro cleavage assay to assess the two cleavage reactions performed on the C-prM precursor. Accordingly, a recombinant modular NS2B/3(pro), consisting of the protease domain of NS3 linked to the core-domain of cofactor NS2B, was expressed in E. coli and purified to homogeneity. This enzyme could cleave a C-prM protein synthesised in rabbit reticulocyte lysates. However, cleavage was only specific when protein synthesis was performed in the presence of canine pancreatic microsomal membranes and required the prevention of signal peptidase I (SPaseI) activity by lengthening the h-region of the signal peptide. The presence of membranes allowed the concentration of NS2B/3(pro) used to be reduced by 10-20 fold. Substitution of the NS2B/3(pro) cleavage motif in C-prM by a HIV-1(pro) motif inhibited NS2B/3(pro) processing in the presence of microsomal membranes but allowed cleavage by HIV-1(pro) at the C-prM junction. This system shows that processing at the C-terminus of protein C by the TBEV NS2B/3(pro) is highly membrane dependent and will allow the examination of how the membrane topology of protein C affects both SPaseI and NS2B/3(pro) processing.


Asunto(s)
Proteínas de la Cápside/metabolismo , Enfermedades de los Perros/virología , Virus de la Encefalitis Transmitidos por Garrapatas/enzimología , Encefalitis Transmitida por Garrapatas/veterinaria , Membranas Intracelulares/virología , Proteínas del Envoltorio Viral/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Enfermedades de los Perros/metabolismo , Perros , Virus de la Encefalitis Transmitidos por Garrapatas/química , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/metabolismo , Encefalitis Transmitida por Garrapatas/metabolismo , Encefalitis Transmitida por Garrapatas/virología , Membranas Intracelulares/metabolismo , Microsomas/metabolismo , Microsomas/virología , Datos de Secuencia Molecular , Proteolisis , ARN Helicasas/química , ARN Helicasas/genética , ARN Helicasas/metabolismo , Alineación de Secuencia , Serina Endopeptidasas/química , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
4.
Eur J Immunol ; 39(1): 85-95, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19089809

RESUMEN

Although matured DC are capable of inducing effective primary and secondary immune responses in vivo, it is difficult to control the maturation and antigen loading in vitro. In this study, we show that ER-enriched microsomal membranes (microsomes) isolated from DC contain more peptide-receptive MHC I and II molecules than, and a similar level of costimulatory molecules to, their parental DC. After loading with defined antigenic peptides, the microsomes deliver antigenic peptide-MHC complexes (pMHC) to both CD4 and CD8 T cells effectively in vivo. The peptide-loaded microsomes accumulate in peripheral lymphoid organs and induce stronger immune responses than peptide-pulsed DC. The microsomal vaccines protect against acute viral infection. Our data demonstrate that peptide-MHC complexes armed microsomes from DC can be an important alternative to DC-based vaccines for protection from viral infection.


Asunto(s)
Presentación de Antígeno , Retículo Endoplásmico/inmunología , Microsomas/inmunología , Ovalbúmina/inmunología , Vacunas Virales/inmunología , Virosis/prevención & control , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/virología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Células Dendríticas/inmunología , Retículo Endoplásmico/virología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microsomas/ultraestructura , Microsomas/virología , Fragmentos de Péptidos/inmunología , Bazo/citología , Bazo/inmunología , Vaccinia/prevención & control
5.
Mol Cell Proteomics ; 8(4): 805-15, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19098277

RESUMEN

In the present work, we have attempted a comprehensive analysis of cytosolic and microsomal proteomes to elucidate the signaling pathways impaired in human hepatoma (Huh7) cells upon herpes simplex virus type 1 (HSV-1; Cgal(+)) infection. Using a combination of differential in-gel electrophoresis and nano liquid chromatography/tandem mass spectrometry, 18 spots corresponding to 16 unique deregulated cellular proteins were unambiguously identified, which were involved in the regulation of essential processes such as apoptosis, mRNA processing, cellular structure and integrity, signal transduction, and endoplasmic-reticulum-associated degradation pathway. Based on our proteomic data and additional functional studies target proteins were identified indicating a late activation of apoptotic pathways in Huh7 cells upon HSV-1 Cgal(+) infection. Additionally to changes on RuvB-like 2 and Bif-1, down-regulation of Erlin-2 suggests stimulation of Ca(2+)-dependent apoptosis. Moreover, activation of the mitochondrial apoptotic pathway results from a time-dependent multi-factorial impairment as inferred from the stepwise characterization of constitutive pro- and anti-apoptotic factors. Activation of serine-threonine protein phosphatase 2A (PP2A) was also found in Huh7 cells upon HSV-1 Cgal(+) infection. In addition, PP2A activation paralleled dephosphorylation and inactivation of downstream mitogen-activated protein (MAP) kinase pathway (MEK(1/2), ERK(1/2)) critical to cell survival and activation of proapoptotic Bad by dephosphorylation of Ser-112. Taken together, our results provide novel molecular information that contributes to define in detail the apoptotic mechanisms triggered by HSV-1 Cgal(+) in the host cell and lead to the implication of PP2A in the transduction of cell death signals and cell survival pathway arrest.


Asunto(s)
Carcinoma Hepatocelular/virología , Herpesvirus Humano 1/fisiología , Neoplasias Hepáticas/virología , Orgánulos/metabolismo , Proteómica , Transducción de Señal , Replicación Viral , Animales , Apoptosis , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Supervivencia Celular , Chlorocebus aethiops , Efecto Citopatogénico Viral , Citosol/metabolismo , Citosol/virología , Activación Enzimática , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Microsomas/metabolismo , Microsomas/virología , Modelos Biológicos , Orgánulos/virología , Proteína Fosfatasa 2/metabolismo , Proteoma/análisis , Proteoma/química , Células Vero
6.
Virus Res ; 137(2): 225-34, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18723056

RESUMEN

Domain III (DIII) of the envelope protein of dengue virus (DENV) contains structural determinants for the interaction with cellular receptors. In the present study a solid phase assay and recombinant fusion proteins containing DENV-DIII of serotypes 1 and 2 were used to study structural features of the interaction of the envelope protein with putative receptors present in the microsomal fraction of CHO cells. Recombinant fusion proteins showed specific interaction with proteins present in the microsomal fraction. Binding of the fusion proteins across the pH range of 5.5-8.0 resembled that of virus particles, peaking at pH 6.0. This suggests that the interaction of DIII with cell receptor(s) is strengthened at endosomal pH. The effect of reduction and carbamidomethylation of cysteine residues on the binding to the microsomal fraction and in their recognition by antibodies suggests that the region of DIII that is interacting with putative receptor(s) overlaps only partially with a dominant epitope of the antibody response. The analysis of the residue conservation profile indicates that the surface of DIII is composed typically of specific sub-complex residues with an increased representation of specific type/subtype residues found at the surface that closely correlates with the dominant neutralizing epitope.


Asunto(s)
Virus del Dengue/metabolismo , Dengue/metabolismo , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Dengue/virología , Virus del Dengue/química , Virus del Dengue/genética , Humanos , Ratones , Microsomas/metabolismo , Microsomas/virología , Unión Proteica , Estructura Terciaria de Proteína , Proteínas del Envoltorio Viral/genética
7.
Diabetes ; 57(7): 1805-13, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18420488

RESUMEN

OBJECTIVE: Human adenovirus type 36 (Ad-36) increases adiposity but improves insulin sensitivity in experimentally infected animals. We determined the ability of Ad-36 to increase glucose uptake by human primary skeletal muscle (HSKM) cells. RESEARCH DESIGN AND METHODS: The effect of Ad-36 on glucose uptake and cell signaling was determined in HSKM cells obtained from type 2 diabetic and healthy lean subjects. Ad-2, another human adenovirus, was used as a negative control. Gene expression and proteins of GLUT1 and GLUT4 were measured by real-time PCR and Western blotting. Role of insulin and Ras signaling pathways was determined in Ad-36-infected HSKM cells. RESULTS: Ad-36 and Ad-2 infections were confirmed by the presence of respective viral mRNA and protein expressions. In a dose-dependent manner, Ad-36 significantly increased glucose uptake in diabetic and nondiabetic HSKM cells. Ad-36 increased gene expression and protein abundance of GLUT1 and GLUT4, GLUT4 translocation to plasma membrane, and phosphatidylinositol 3-kinase (PI 3-kinase) activity in an insulin-independent manner. In fact, Ad-36 decreased insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation and IRS-1-and IRS-2-associated PI 3-kinase activities. On the other hand, Ad-36 increased Ras gene expression and protein abundance, and Ras siRNA abrogated Ad-36-induced PI 3-kinase activation, GLUT4 protein abundance, and glucose uptake. These effects were not observed with Ad-2 infection. CONCLUSIONS: Ad-36 infection increases glucose uptake in HSKM cells via Ras-activated PI 3-kinase pathway in an insulin-independent manner. These findings may provide impetus to exploit the role of Ad-36 proteins as novel therapeutic targets for improving glucose handling.


Asunto(s)
Infecciones por Adenovirus Humanos/metabolismo , Adenovirus Humanos/fisiología , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Insulina/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/virología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Adenovirus Humanos/fisiopatología , Membrana Celular/metabolismo , Membrana Celular/virología , Desoxiglucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microsomas/metabolismo , Microsomas/virología , Músculo Esquelético/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transfección
8.
J Virol ; 78(8): 4299-1313, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047843

RESUMEN

The assembly of African swine fever virus (ASFV) at the cytoplasmic virus factories commences with the formation of precursor membranous structures, which are thought to be collapsed cisternal domains recruited from the surrounding endoplasmic reticulum (ER). This report analyzes the role in virus morphogenesis of the structural protein p54, a 25-kDa polypeptide encoded by the E183L gene that contains a putative transmembrane domain and localizes at the ER-derived envelope precursors. We show that protein p54 behaves in vitro and in infected cells as a type I membrane-anchored protein that forms disulfide-linked homodimers through its unique luminal cysteine. Moreover, p54 is targeted to the ER membranes when it is transiently expressed in transfected cells. Using a lethal conditional recombinant, vE183Li, we also demonstrate that the repression of p54 synthesis arrests virus morphogenesis at a very early stage, even prior to the formation of the precursor membranes. Under restrictive conditions, the virus factories appeared as discrete electron-lucent areas essentially free of viral structures. In contrast, outside the assembly sites, large amounts of aberrant zipper-like structures formed by the unprocessed core polyproteins pp220 and pp62 were produced in close association to ER cisternae. Altogether, these results indicate that the transmembrane structural protein p54 is critical for the recruitment and transformation of the ER membranes into the precursors of the viral envelope.


Asunto(s)
Virus de la Fiebre Porcina Africana/fisiología , Productos del Gen env/fisiología , Proteínas Estructurales Virales/fisiología , Virus de la Fiebre Porcina Africana/genética , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , ADN Viral/genética , Dimerización , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Expresión Génica , Productos del Gen env/genética , Genes Virales , Microscopía Electrónica , Microsomas/metabolismo , Microsomas/virología , Precursores de Proteínas/genética , Precursores de Proteínas/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Células Vero , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/genética , Ensamble de Virus
9.
Virology ; 288(1): 89-95, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11543661

RESUMEN

Many glycoproteins of enveloped viruses as well as cellular proteins are covalently modified with fatty acids. Palmitoylation is often reversible, but the enzymology of this hydrophobic protein modification is not understood. Recently a cytosolic enzyme designated acyl-protein thioesterase 1 (APT1) was purified, which depalmitoylates several cellular proteins. Since hitherto no transmembrane proteins have been tested as substrates for APT1 we have investigated whether palmitoylated viral membrane glycoproteins can be deacylated by use of this enzyme. Recombinant APT1 was purified from Escherichia coli, and depalmitoylation of [3H]palmitate-labeled glycoproteins present in virus particles was measured by SDS-PAGE, fluorography, and scanning densitometry. We find that APT1 causes rapid and almost complete cleavage of fatty acids from the G-protein of vesicular stomatitis virus, hemagglutinin proteins of influenza A and C virus, and E2 of Semliki Forest virus (SFV). In contrast, E1 of SFV is largely resistant against APT1 activity. This substrate specificity of APT1 was also observed using microsomes prepared from SFV-infected cells. Our data emphasize the potential of APT1 as a tool for functional analysis of protein-bound fatty acids.


Asunto(s)
Glicoproteínas/metabolismo , Virus de la Influenza A/fisiología , Ácido Palmítico/metabolismo , Virus de los Bosques Semliki/fisiología , Tioléster Hidrolasas/metabolismo , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas Virales/metabolismo , Animales , Línea Celular , Cricetinae , Citosol/enzimología , Perros , Escherichia coli/enzimología , Riñón , Microsomas/virología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Tioléster Hidrolasas/aislamiento & purificación
10.
Yeast ; 13(4): 299-304, 1997 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-9133733

RESUMEN

The microsomal fraction isolated form serine palmitoyltransferase (lcb2/scs1) mutants is enriched in a 90 kDa protein. The protein was identified as the major coat (Gag) protein of the L-A dsRNA virus particles by partial sequencing and by its interaction with anti-Gag antibodies. The total amount of Gag in whole-cell lysates of scs1/lcb2 mutant cells is greater than in wild-type lysates indicating that the enrichment of the protein in the microsomal fraction of scs1/lcb2 mutant cells may result from increased copy number of the L-A dsRNA virus. This is supported by the findings that the mutants also have increased levels of L-A dsRNA. Altered sphingolipid synthesis in the scs1 mutant cells appears to increase the copy number of the L-A viral particles.


Asunto(s)
Aciltransferasas/genética , Virus ARN/aislamiento & purificación , ARN Bicatenario/aislamiento & purificación , ARN Viral/aislamiento & purificación , Saccharomyces cerevisiae/virología , Secuencia de Aminoácidos , Anticuerpos Antivirales , Cápside/análisis , Cápside/química , Cápside/genética , Productos del Gen gag/análisis , Productos del Gen gag/química , Productos del Gen gag/genética , Microsomas/virología , Datos de Secuencia Molecular , Peso Molecular , Mutación/fisiología , Fragmentos de Péptidos/química , Virus ARN/inmunología , Proteínas Recombinantes de Fusión , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae , Análisis de Secuencia , Serina C-Palmitoiltransferasa , Esfingolípidos/biosíntesis
11.
Virology ; 227(1): 245-51, 1997 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-9007081

RESUMEN

Open reading frame (ORF) 3 of the genome of lactate dehydrogenase-elevating virus (LDV), strain P, was cloned into the plasmid pcDNAI/Amp and in vitro transcribed and translated. Translation of ORF 3 yielded a soluble protein of the expected size (about 21 kDa). When synthesized in the presence of endoplasmic reticulum (ER) membranes the resulting glycoprotein of about 36 kDa became associated with the membranes. However, disruption of the ER vesicles by incubation in carbonate buffer, pH 11.5, resulted in the release of the protein from the membranes. Hydrophobic moment analysis of the ORF 3 protein indicated the absence of any potential transmembrane segments, except for a N-terminal signal peptide, but no cleavage of the signal peptide was observed during membrane-associated in vitro synthesis. The ORF 3 protein elicited a strong antibody response in infected mice. The antibodies from infected mice as well as a monoclonal antibody specifically precipitated the in vitro-synthesized ORF 3 protein, but no protein from LDV virions. The overall results suggest that the ORF 3 protein is a nonstructural, highly glycosylated, and antigenic glycoprotein that is probably soluble and secreted or at most only weakly associated with membranes via the signal peptide.


Asunto(s)
Virus Elevador de Lactato Deshidrogenasa/genética , Sistemas de Lectura Abierta , Proteínas no Estructurales Virales/genética , Animales , Anticuerpos Antivirales/inmunología , Infecciones por Arterivirus/inmunología , Secuencia de Bases , Células COS , Células Cultivadas , Clonación Molecular , ADN Viral , Membranas Intracelulares/virología , Virus Elevador de Lactato Deshidrogenasa/inmunología , Macrófagos/virología , Ratones , Microsomas/virología , Datos de Secuencia Molecular , Señales de Clasificación de Proteína/metabolismo , Proteínas no Estructurales Virales/inmunología
12.
Arch Virol ; 141(10): 2009-17, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8920832

RESUMEN

The intracellular localization of the glycoprotein B of herpesvirus simian agent 8 expressed with a baculovirus system in insect cells was studied. Cell fractionation and immunoprecipitation revealed that gB is present in microsomal as well as in nuclear membranes. Both fractions contain oligomers, probably dimers, of gB with endoglycosidase-H sensitive, mannose-rich carbohydrates. Nuclear transport of gB was further analysed by immuno electron microscopy of recombinant baculovirus-infected cells. The glycoprotein is present both in the outer and the inner nuclear membrane as well as in cytoplasmic structures and at the cell surface. This study precludes the possibility that glycosylation and/or oligomerisation of SA8 gB are responsible for nuclear targeting.


Asunto(s)
Herpesvirus Cercopitecino 1/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Baculoviridae/genética , Línea Celular , Expresión Génica , Vectores Genéticos , Microsomas/metabolismo , Microsomas/virología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Spodoptera/citología , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/análisis
13.
J Virol ; 68(12): 7859-68, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7966576

RESUMEN

The molecular basis of strain variation in scrapie diseases is unknown. The only identified component of the agent is the posttranslationally modified host prion protein (PrPSc). The biochemical and physical properties of PrP from two strains of transmissible mink encephalopathy (TME), called hyper (HY) and drowsy (DY), were compared to investigate if PrP heterogeneity could account for strain diversity. The degradation rate of PrPTME digested with proteinase K was found to be strain specific and correlated with inactivation of the TME titer. Edman protein sequencing revealed that the major N-terminal end of HY PrPTME commenced at least 10 amino acid residues prior to that of DY PrPTME after digestion with proteinase K. Analysis of the brain distribution of PrPTME exhibited a strain-specific pattern and localization of PrPTME to the perikarya of specific neuron populations. Our findings are consistent with HY and DY PrPTME having distinct protein conformations and/or strain-specific ligand interactions that influence PrPTME properties. We propose that PrPTME conformation could play a role in targeting TME strains to different neuron populations in which strain-specific formation occurs. These data are consistent with the idea that PrPTME protein structure determines the molecular basis of strain variation.


Asunto(s)
Encéfalo/virología , Variación Genética , Microsomas/virología , Enfermedades por Prión/patología , Priones/genética , Secuencia de Aminoácidos , Animales , Encéfalo/patología , Cricetinae , Masculino , Mesocricetus , Microsomas/patología , Visón , Datos de Secuencia Molecular , Especificidad de Órganos , Enfermedades por Prión/virología , Priones/aislamiento & purificación , Priones/patogenicidad , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...