Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ann Neurol ; 94(3): 531-546, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37190821

RESUMEN

OBJECTIVE: Peroxisome injury occurs in the central nervous system (CNS) during multiple virus infections that result in neurological disabilities. We investigated host neuroimmune responses and peroxisome biogenesis factors during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection using a multiplatform strategy. METHODS: Brain tissues from coronavirus disease 2019 (COVID-19) (n = 12) and other disease control (ODC) (n = 12) patients, as well as primary human neural cells and Syrian hamsters, infected with a clinical variant of SARS-CoV-2, were investigated by droplet digital polymerase chain reaction (ddPCR), quantitative reverse transcriptase PCR (RT-qPCR), and immunodetection methods. RESULTS: SARS-CoV-2 RNA was detected in the CNS of 4 patients with COVID-19 with viral protein (NSP3 and spike) immunodetection in the brainstem. Olfactory bulb, brainstem, and cerebrum from patients with COVID-19 showed induction of pro-inflammatory transcripts (IL8, IL18, CXCL10, NOD2) and cytokines (GM-CSF and IL-18) compared to CNS tissues from ODC patients (p < 0.05). Peroxisome biogenesis factor transcripts (PEX3, PEX5L, PEX11ß, and PEX14) and proteins (PEX3, PEX14, PMP70) were suppressed in the CNS of COVID-19 compared to ODC patients (p < 0.05). SARS-CoV-2 infection of hamsters revealed viral RNA detection in the olfactory bulb at days 4 and 7 post-infection while inflammatory gene expression was upregulated in the cerebrum of infected animals by day 14 post-infection (p < 0.05). Pex3 transcript levels together with catalase and PMP70 immunoreactivity were suppressed in the cerebrum of SARS-CoV-2 infected animals (p < 0.05). INTERPRETATION: COVID-19 induced sustained neuroinflammatory responses with peroxisome biogenesis factor suppression despite limited brainstem SARS-CoV-2 neurotropism in humans. These observations offer insights into developing biomarkers and therapies, while also implicating persistent peroxisome dysfunction as a contributor to the neurological post-acute sequelae of COVID-19. ANN NEUROL 2023;94:531-546.


Asunto(s)
COVID-19 , Animales , Humanos , SARS-CoV-2 , Enfermedades Neuroinflamatorias , ARN Viral , Peroxisomas , Encéfalo
2.
J Virol ; 95(23): e0107421, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34524914

RESUMEN

Human pegivirus (HPgV) infects peripheral leukocytes but was recently shown to be a neurotropic virus associated with leukoencephalitis in humans. In the present study, we investigated the neural cell tropism of HPgV as well as its effects on host immune responses. HPgV wild type (WT) and a mutant virus with a deletion in the HPgV NS2 gene (ΔNS2) were able to productively infect human astrocytes and microglia but not neurons or an oligodendrocyte-derived cell line. Of note, the ΔNS2 virus replicated better than WT pegivirus in astrocytes, with both viruses being able to subsequently infect and spread in fresh human astrocyte cultures. Infection of human glia by HPgV WT and ΔNS2 viruses resulted in suppression of peroxisome-associated genes, including PEX11B, ABCD1, PEX7, ABCD3, PEX3, and PEX5L, during peak viral production, which was accompanied by reduced expression of IFNB, IRF3, IRF1, and MAVS, particularly in ΔNS2-infected cells. These data were consistent with analyses of brain tissue from patients infected with HPgV in which we observed suppression of peroxisome and type I interferon gene transcripts, including PEX11B, ABCD3, IRF1, and IRF3, with concurrent loss of PMP70 immunoreactivity in glia. Our data indicate that human astrocytes and microglia are permissive to HPgV infection, resulting in peroxisome injury and suppressed antiviral signaling that is influenced by viral diversity. IMPORTANCE Human pegiviruses are detected in 1 to 5% of the general population, principally infecting leukocytes, although their effects on human health remain uncertain. Here, we show that human pegivirus infects specific neural cell types in culture and human brain and, like other neurotropic flaviviruses, causes suppression of peroxisome and antiviral signaling pathways, which could favor ongoing viral infection and perhaps confer susceptibility to the development of neurological disease.


Asunto(s)
Antivirales/farmacología , Infecciones por Flaviviridae/metabolismo , Neuroglía/metabolismo , Pegivirus/metabolismo , Transducción de Señal/efectos de los fármacos , Astrocitos , Encéfalo/metabolismo , Encéfalo/patología , Infecciones por Flaviviridae/genética , Infecciones por Flaviviridae/virología , Expresión Génica , Humanos , Microglía/metabolismo , Microglía/virología , Neuroglía/patología , Neuroglía/virología , Pegivirus/efectos de los fármacos , Pegivirus/genética , Filogenia , ARN Viral/genética , Proteínas no Estructurales Virales/genética
3.
J Biol Chem ; 276(46): 43294-9, 2001 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-11553639

RESUMEN

GERp95 (Golgi-endoplasmic reticulum protein 95 kDa) is part of a large family of highly conserved proteins found in all metazoans and the fission yeast Schizosaccharomyces pombe. Genetic studies suggest that homologs of GERp95 are components of signaling pathways that regulate cellular differentiation, development, and RNA interference. However, the precise molecular functions of these proteins remain unknown. Genetic analysis of GERp95 homologs has been complicated by the presence of multiple genes with overlapping functions in most organisms. Binding partners for members of this protein family have not been identified. The purpose of this study was to identify proteins that associate with GERp95. Glutathione S-transferase-GERp95 fusions were expressed in transfected cells, and proteins that bound to GERp95 were co-purified using glutathione-agarose beads. The amino-terminal region of GERp95 was found to interact with the specialized chaperone Hsp90 and a number of its cognate binding proteins. Inhibition of Hsp90 activity with geldanamycin or radicicol resulted in rapid degradation of newly synthesized GERp95. The membrane-associated pool of GERp95 was not bound to Hsp90, although activity of this chaperone was required for stable association of GERp95 with the Golgi in normal rat kidney cells. These results indicate that GERp95 engages an Hsp90 chaperone complex prior to association with intracellular membranes.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Transducción de Señal , Animales , Proteínas Argonautas , Benzoquinonas , Células COS , Línea Celular , Membrana Celular/metabolismo , Electroforesis en Gel de Poliacrilamida , Factor 2 Eucariótico de Iniciación , Regulación de la Expresión Génica , Biblioteca de Genes , Glutatión Transferasa/metabolismo , Humanos , Riñón/citología , Lactamas Macrocíclicas , Microscopía Fluorescente , Pruebas de Precipitina , Unión Proteica , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Quinonas/farmacología , Conejos , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Reticulocitos/metabolismo , Transcripción Genética , Transfección , Técnicas del Sistema de Dos Híbridos
4.
Arch Virol ; 146(1): 1-14, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11266204

RESUMEN

Very little is known about the cellular factors that are required for the maturation of rubella virus glycoproteins (E2 and E1) in the endoplasmic reticulum of the infected cell. In the present study, we established the interaction of the ER chaperone proteins, calreticulin and calnexin, with the RV E1 and E2 proteins in cells stably expressing the viral proteins. The interaction between E2 and calnexin was significantly higher than with calreticulin. In pulse-chase experiments, the half-life of the E2-calnexin was >45 min, whereas the half-life of the calreticulin-E2 interaction was approximately 10 min. Tunicamycin and castanospermine treatments altered the mobilities of intracellular E1 and E2, due to either lack of oligosaccharide ligand addition or trimming of terminal glucose residues, respectively. Further, the drug treatments resulted in a loss of E1 and E2 interaction with calreticulin or calnexin, thereby demonstrating that the interaction is through monoglucosylated forms of RV proteins. These studies suggest that the interaction of RV glycoproteins with the ER chaperone proteins is essential for their maturation in the endoplasmic reticulum.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Glicoproteínas/metabolismo , Chaperonas Moleculares/metabolismo , Ribonucleoproteínas/metabolismo , Virus de la Rubéola/metabolismo , Animales , Antivirales/farmacología , Células CHO/efectos de los fármacos , Células CHO/virología , Calnexina , Calreticulina , Cricetinae , Retículo Endoplásmico/virología , Inhibidores Enzimáticos/farmacología , Indolizinas/farmacología , Cinética , Unión Proteica , Tunicamicina/farmacología , Proteínas del Envoltorio Viral/metabolismo
5.
J Virol ; 75(4): 1978-83, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11160697

RESUMEN

The rubella virus (RV) structural proteins capsid, E2, and E1 are synthesized as a polyprotein precursor. The signal peptide that initiates translocation of E2 into the lumen of the endoplasmic reticulum remains attached to the carboxy terminus of the capsid protein after cleavage by signal peptidase. Among togaviruses, this feature is unique to RV. The E2 signal peptide has previously been shown to function as a membrane anchor for the capsid protein. In the present study, we demonstrate that this domain is required for RV glycoprotein-dependent localization of the capsid protein to the juxtanuclear region and subsequent virus assembly at the Golgi complex.


Asunto(s)
Cápside/metabolismo , Señales de Clasificación de Proteína/fisiología , Virus de la Rubéola/fisiología , Proteínas del Envoltorio Viral/metabolismo , Ensamble de Virus , Animales , Células COS , Cápside/genética , Señales de Clasificación de Proteína/genética , Transporte de Proteínas , Virus de la Rubéola/genética , Virus de la Rubéola/metabolismo , Transfección , Proteínas del Envoltorio Viral/genética
6.
Virology ; 275(1): 20-9, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11017784

RESUMEN

Rubella virus is an enveloped positive-strand RNA virus that can cause mild to severe birth defects or death in an infected fetus. RV induction of programmed cell death, demonstrated in cell culture, has been implicated in the pathogenesis. The timing of apoptosis, 48 h p.i., suggested that accumulation of RV structural proteins might induce cell death in infected cells. Expression of RV structural proteins, capsid, envelope glycoproteins E1 and E2, in transiently transfected RK13 cells was as potent an inducer of cell death as RV infection. Immunofluorescence microscopy revealed that RV structural protein transfected cells exhibited the condensed nuclei typical of apoptotic cell death. Transfection with the capsid protein construct, but not E2 and E1, resulted in as much cell death as joint expression of all three RV structural proteins. Capsid required a membrane-anchoring domain to induce cell death, but a heterologous polypeptide fused to the capsid membrane anchor did not cause apoptosis. Deletion mutants demonstrated that the apoptosis-inducing activity resides in the N-terminal 170 amino acids of capsid. Though apoptosis-inducing capsid constructs appear to have an ER sub-cellular localization, disruption of the ER calcium storage capacity does not correlate with cell death. Mechanisms consistent with these results are discussed.


Asunto(s)
Apoptosis , Cápside/fisiología , Virus de la Rubéola/fisiología , Animales , Apoptosis/efectos de los fármacos , Transporte Biológico , Western Blotting , Calcio/metabolismo , Cápside/genética , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Etiquetado Corte-Fin in Situ , Ratones , Señales de Clasificación de Proteína/genética , Señales de Clasificación de Proteína/fisiología , Conejos , Virus de la Rubéola/genética , Eliminación de Secuencia/genética , Tapsigargina/farmacología , Transfección
7.
J Virol ; 74(12): 5569-76, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10823864

RESUMEN

Togavirus nucleocapsids have a characteristic icosahedral structure and are composed of multiple copies of a capsid protein complexed with genomic RNA. The assembly of rubella virus nucleocapsids is unique among togaviruses in that the process occurs late in virus assembly and in association with intracellular membranes. The goal of this study was to identify host cell proteins which may be involved in regulating rubella virus nucleocapsid assembly through their interactions with the capsid protein. Capsid was used as bait to screen a CV1 cDNA library using the yeast two-hybrid system. One protein that interacted strongly with capsid was p32, a cellular protein which is known to interact with other viral proteins. The interaction between capsid and p32 was confirmed using a number of different in vitro and in vivo methods, and the site of interaction between these two proteins was shown to be at the mitochondria. Interestingly, overexpression of the rubella virus structural proteins resulted in clustering of the mitochondria in the perinuclear region. The p32-binding site in capsid is a potentially phosphorylated region that overlaps the viral RNA-binding domain of capsid. Our results are consistent with the possibility that the interaction of p32 with capsid plays a role in the regulation of nucleocapsid assembly and/or virus-host interactions.


Asunto(s)
Cápside/metabolismo , Receptores de Hialuranos , Mitocondrias/metabolismo , Proteínas/química , Proteínas/metabolismo , Virus de la Rubéola , Animales , Sitios de Unión , Cápside/química , Cápside/genética , Línea Celular , Chlorocebus aethiops , Citoplasma/química , Técnica del Anticuerpo Fluorescente Indirecta , Mitocondrias/química , Datos de Secuencia Molecular , Peso Molecular , Fosforilación , Pruebas de Precipitina , Unión Proteica , Proteínas/genética , Virus de la Rubéola/genética , Eliminación de Secuencia/genética , Especificidad por Sustrato , Transfección , Técnicas del Sistema de Dos Híbridos
8.
Mol Biol Cell ; 10(10): 3357-72, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10512872

RESUMEN

A panel of mAbs was elicited against intracellular membrane fractions from rat pancreas. One of the antibodies reacted with a 95-kDa protein that localizes primarily to the Golgi complex or the endoplasmic reticulum (ER), depending on cell type. The corresponding cDNA was cloned and sequenced and found to encode a protein of 97.6 kDa that we call GERp95 (Golgi ER protein 95 kDa). The protein copurifies with intracellular membranes but does not contain hydrophobic regions that could function as signal peptides or transmembrane domains. Biochemical analysis suggests that GERp95 is a cytoplasmically exposed peripheral membrane protein that exists in a protease-resistant complex. GERp95 belongs to a family of highly conserved proteins in metazoans and Schizosaccharomyces pombe. It has recently been determined that plant and Drosophila homologues of GERp95 are important for controlling the differentiation of stem cells (Bohmert et al., 1998; Cox et al., 1998; Moussian et al., 1998). In Caenorhabditis elegans, there are at least 20 members of this protein family. To this end, we have used RNA interference to show that the GERp95 orthologue in C. elegans is important for maturation of germ-line stem cells in the gonad. GERp95 and related proteins are an emerging new family of proteins that have important roles in metazoan development. The present study suggests that these proteins may exert their effects on cell differentiation from the level of intracellular membranes.


Asunto(s)
Diferenciación Celular , Proteínas de la Membrana/genética , Páncreas/química , Células Madre/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Argonautas , Células COS , Caenorhabditis elegans/metabolismo , Clonación Molecular , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación , Técnica del Anticuerpo Fluorescente , Células Germinativas/metabolismo , Aparato de Golgi/metabolismo , Membranas Intracelulares/química , Proteínas de la Membrana/química , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Proteínas Recombinantes , Reproducción , Alineación de Secuencia , Células Madre/citología , Transfección
9.
Cancer Res ; 59(19): 4890-7, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10519401

RESUMEN

Mouse transporter protein (MTP), a small, highly conserved mammalian intracellular membrane protein with four putative transmembrane domains, has been implicated in the transport of nucleosides and/or related molecules across intracellular membranes. The production of recombinant MTP in Saccharomyces cerevisiae alters sensitivity of yeast cells to a heterogeneous group of compounds (e.g., antimetabolites, antibiotics, anthracyclines, ionophores, and steroid hormones) by changing the subcellular compartmentalization of these drugs, suggesting that MTP functions similarly in higher organisms. The present study was undertaken to define the intracellular location of MTP in mammalian cells. Native MTP was not detected by indirect immunofluorescence in cell types that expressed MTP mRNA; therefore, a hemagglutinin (HA) epitope-tagged version of MTP was produced in cultured BHK21 cells by transient transfection, and its distribution within cells was determined by confocal microscopy using antibodies directed against the HA epitope and various organellar proteins. Antibodies directed against HA-MTP colocalized with antibodies against late endosomal and lysosomal proteins but not with antibodies against either Golgi or early endosomal proteins. Analysis of subcellular fractions from rat liver by immunoblotting with antibodies directed against MTP demonstrated the presence of a MTP-like protein in Golgi- and lysosome-enriched membranes but not in mitochondria. These results indicate that MTP resides in late endosomes and lysosomes, a finding that is consistent with the proposed role for MTP in the movement of a variety of small molecules across endosomal and lysosomal membranes. MTP shares a number of characteristics with other lysosome-associated proteins. We, therefore, propose that it be redesignated murine lysosome-associated protein transmembrane 4.


Asunto(s)
Proteínas Portadoras/metabolismo , Resistencia a Múltiples Medicamentos , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Fraccionamiento Celular/métodos , Línea Celular , Cricetinae , Endosomas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Aparato de Golgi/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Nucleósidos/metabolismo , Ratas , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección
10.
Virology ; 261(2): 340-6, 1999 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10497119

RESUMEN

Rubella virus (RV) is an enveloped RNA virus that causes systemic infections in humans. More importantly, first trimester in utero infection leads to a collection of devastating birth defects known as congenital rubella syndrome. Epithelial cells are the first line of defense against viruses and consequently, the polarity of virus secretion is an important factor affecting viral spread. As a first step toward understanding how RV interacts with epithelial cells, we have examined the release of RV-like particles and virions from polarized cells in culture. RV structural proteins were targeted to the Golgi complex and virus particle formation occurred on intracellular membranes in three different polarized epithelial cells. Polarized cells could be infected from the apical and basal membranes, indicating that receptors are not confined to one surface. The secretion of virus-like particles and infectious virions varied according to cell type. In two of the three polarized cell lines examined, virus was released primarily from the apical surface, but significant quantities were also secreted from the basolateral membrane. Release of virus from the apical surface may facilitate virus spread from person to person, whereas basolateral secretion could be important for establishing a systemic infection and/or crossing the placenta prior to fetal infection.


Asunto(s)
Células Epiteliales/virología , Virus de la Rubéola/fisiología , Rubéola (Sarampión Alemán)/virología , Virión/fisiología , Replicación Viral , Línea Celular , Humanos , Rubéola (Sarampión Alemán)/patología , Ensamble de Virus
11.
J Virol ; 73(5): 3524-33, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10196241

RESUMEN

Rubella virus is a small enveloped positive-strand RNA virus that assembles on intracellular membranes in a variety of cell types. The virus structural proteins contain all of the information necessary to mediate the assembly of virus-like particles in the Golgi complex. We have recently identified intracellular retention signals within the two viral envelope glycoproteins. E2 contains a Golgi retention signal in its transmembrane domain, whereas a signal for retention in the endoplasmic reticulum has been localized to the transmembrane and cytoplasmic domains of E1 (T. C. Hobman, L. Woodward, and M. G. Farquhar, Mol. Biol. Cell 6:7-20, 1995; T. C. Hobman, H. F. Lemon, and K. Jewell, J. Virol. 71:7670-7680, 1997). In the present study, we have analyzed the role of these retention signals in the assembly of rubella virus-like particles. Deletion or replacement of these domains with analogous regions from other type I membrane glycoproteins resulted in failure of rubella virus-like particles to be secreted from transfected cells. The E1 transmembrane and cytoplasmic domains were not required for targeting of the structural proteins to the Golgi complex and, surprisingly, assembly and budding of virus particles into the lumen of this organelle; however, the resultant particles were not secreted. In contrast, replacement or alteration of the E2 transmembrane or cytoplasmic domain, respectively, abrogated the targeting of the structural proteins to the budding site, and consequently, no virion formation was observed. These results indicate that the transmembrane and cytoplasmic domains of E2 and E1 are required for early and late steps respectively in the viral assembly pathway and that rubella virus morphogenesis is very different from that of the structurally similar alphaviruses.


Asunto(s)
Glicoproteínas/fisiología , Virus de la Rubéola/fisiología , Proteínas del Envoltorio Viral/fisiología , Ensamble de Virus , Animales , Sitios de Unión , Transporte Biológico , Dimerización , Glicoproteínas/genética , Glicoproteínas/metabolismo , Aparato de Golgi/ultraestructura , Aparato de Golgi/virología , Humanos , Virus de la Rubéola/metabolismo , Virus de la Rubéola/ultraestructura , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virión/fisiología , Virión/ultraestructura
12.
Mol Biol Cell ; 9(6): 1265-78, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9614173

RESUMEN

Rubella virus E1 glycoprotein normally complexes with E2 in the endoplasmic reticulum (ER) to form a heterodimer that is transported to and retained in the Golgi complex. In a previous study, we showed that in the absence of E2, unassembled E1 subunits accumulate in a tubular pre-Golgi compartment whose morphology and biochemical properties are distinct from both rough ER and Golgi. We hypothesized that this compartment corresponds to hypertrophied ER exit sites that have expanded in response to overexpression of E1. In the present study we constructed BHK cells stably expressing E1 protein containing a cytoplasmically disposed epitope and isolated the pre-Golgi compartment from these cells by cell fractionation and immunoisolation. Double label indirect immunofluorescence in cells and immunoblotting of immunoisolated tubular networks revealed that proteins involved in formation of ER-derived transport vesicles, namely p58/ERGIC 53, Sec23p, and Sec13p, were concentrated in the E1-containing pre-Golgi compartment. Furthermore, budding structures were evident in these membrane profiles, and a highly abundant but unknown 65-kDa protein was also present. By comparison, marker proteins of the rough ER, Golgi, and COPI vesicles were not enriched in these membranes. These results demonstrate that the composition of the tubular networks corresponds to that expected of ER exit sites. Accordingly, we propose the name SEREC (smooth ER exit compartment) for this structure.


Asunto(s)
Retículo Endoplásmico Rugoso/metabolismo , Proteínas Fúngicas/metabolismo , Lectinas de Unión a Manosa , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae , Proteínas del Envoltorio Viral/metabolismo , Animales , Sitios de Unión , Vesículas Cubiertas por Proteínas de Revestimiento , Línea Celular , Vesículas Cubiertas/metabolismo , Cricetinae , Proteínas Activadoras de GTPasa , Aparato de Golgi/metabolismo , Membranas Intracelulares/metabolismo , Mutagénesis , Proteínas de Complejo Poro Nuclear , Transfección , Proteínas del Envoltorio Viral/genética
13.
J Virol ; 71(10): 7670-80, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9311850

RESUMEN

Rubella virus contains three structural proteins, capsid, E2, and E1. E2 and E1 are type I membrane glycoproteins that form a heterodimer in the endoplasmic reticulum (ER) before they are transported to and retained in the Golgi complex, where virus assembly occurs. The bulk of unassembled E2 and E1 subunits are not transported to the Golgi complex. We have recently shown that E2 contains a Golgi-targeting signal that mediates retention of the E2-E1 complex (T. C. Hobman, L. Woodward, and M. G. Farquhar, Mol. Biol. Cell 6:7-20, 1995). The focus of this study was to determine if E1 glycoprotein also contains intracellular targeting information. We constructed a series of chimeric reporter proteins by fusing domains from E1 to the ectodomains of two other type I membrane proteins which are normally transported to the cell surface, vesicular stomatitis virus G protein (G) and CD8. Fusion of the E1 transmembrane and cytoplasmic regions, but not analogous domains from two control membrane proteins, to the ectodomains of G and CD8 proteins caused the resulting chimeras to be retained in the ER. Association of the ER-retained chimeras with known ER chaperone proteins was not detected. ER localization required both the transmembrane and cytoplasmic regions of E1, since neither of these domains alone was sufficient to retain the reporter proteins. Increasing the length of the E1 cytoplasmic domain by 10 amino acids completely abrogated ER retention. This finding also indicated that the chimeras were not retained as a result of misfolding. In summary, we have identified a new type of ER retention signal that may function to prevent unassembled E1 subunits and/or immature E2-E1 dimers from reaching the Golgi complex, where they could interfere with viral assembly. Accordingly, assembly of E2 and E1 would mask the signal, thereby allowing transport of the heterodimer from the ER.


Asunto(s)
Retículo Endoplásmico/virología , Virus de la Rubéola/fisiología , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Antígenos CD8/química , Células CHO , Cápside/metabolismo , Línea Celular , Cricetinae , Cartilla de ADN , Dimerización , Retículo Endoplásmico/fisiología , Genes Reporteros , Fusión de Membrana , Modelos Estructurales , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Proteínas del Envoltorio Viral/química , Replicación Viral
14.
Clin Diagn Virol ; 5(1): 13-20, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15566856

RESUMEN

BACKGROUND: We have investigated the performance of the novel rubella serology assay, Cobas Core Rubella IgG EIA recomb, which uses rubella-like particles (RLPs) expressed in transfected BHK-21 cells as the antigen. STUDY DESIGN: Evaluation of the assay included comparison with the hemagglutination inhibition (HAI) assay and another enzyme immunoassay (EIA) using native rubella virus (RV) as antigen, i.e. the Abbott IMx Rubella IgG. The assay was calibrated against the WHO 1000 IU/ml reference serum and showed good correlation with the HAI test in the analysis of 404 serum samples. However, quantitative differences in IgG values measured in the Cobas Core and the Abbott IMx assays were noted. RESULTS: Values obtained for patient sera as well as CDC and WHO standards were generally more than twice as high in the Abbott IMx assay as in the Cobas Core test. CONCLUSIONS: For sera whose IgG levels in the immunoassay and HAI test were discordant, immunoblotting proved valuable as a confirmatory method and indicated that a significant number of HAI-negative samples were correctly interpreted as positive by the immunoassay.

15.
J Biol Chem ; 270(42): 24924-31, 1995 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-7559618

RESUMEN

The site of apolipoprotein B (apoB) degradation was investigated in cultured rat hepatocytes. Brefeldin A plus nocodazole completely blocked apoB degradation suggesting the involvement of a post-endoplasmic reticulum (ER) compartment. Monensin inhibited apoB degradation by 40% implying that a post-Golgi compartment could be involved in degradation of apoB. Ammonium chloride or chloroquine inhibited partially the degradation of apoB100 and apoB48, indicating some degradation in lysosomes, or in an acidic compartment such as trans-Golgi or endosomes. The degradations of apoB100 and apoB48 were blocked completely by (2S,3S)-trans-epoxysuccinyl-L-leucylamido-3-methylbutane ethyl ester (EST) during a chase of 90 min demonstrating that a cysteine protease was responsible for apoB degradation. Chymostatin, leupeptin, pepstatin, phenylmethylsulfonyl fluoride, and aprotinin had no significant effect on the degradation of apoB48. However, leupeptin and pepstatin decreased the degradation of apoB100 by 20-30%. Degradation of apoB100 and apoB48 occurred in isolated Golgi fractions with little degradation in heavy or light ER. Degradation of apoB in Golgi fractions was inhibited by EST and by preincubating hepatocytes with 10 nM dexamethasone. Immunofluorescent microscopy revealed that apoB accumulated in the Golgi region after EST treatment. It is concluded that a major part of apoB degradation in rat hepatocytes occurs in a post-ER compartment via the action of a cysteine protease that is regulated by glucocorticoids.


Asunto(s)
Apolipoproteínas B/metabolismo , Retículo Endoplásmico/metabolismo , Hígado/metabolismo , Animales , Apolipoproteína B-100 , Células Cultivadas , Dexametasona/farmacología , Aparato de Golgi/metabolismo , Leucina/análogos & derivados , Leucina/farmacología , Lipoproteínas VLDL/metabolismo , Hígado/ultraestructura , Masculino , Ratas , Ratas Sprague-Dawley
16.
Mol Biol Cell ; 6(1): 7-20, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7749196

RESUMEN

Rubella virus (RV) envelope glycoproteins, E2 and E1, form a heterodimeric complex that is targeted to medial/trans-Golgi cisternae. To identify the Golgi targeting signal(s) for the E2/E1 spike complex, we constructed chimeric proteins consisting of domains from RV glycoproteins and vesicular stomatitis virus (VSV) G protein. The location of the chimeric proteins in stably transfected Chinese hamster ovary cells was determined by immunofluorescence, immunoelectron microscopy, and by the extent of processing of their N-linked glycans. A trans-dominant Golgi retention signal was identified within the C-terminal region of E2. When the transmembrane (TM) and cytoplasmic (CT) domains of VSV G were replaced with those of RV E2, the hybrid protein (G-E2TMCT+) was retained in the Golgi. Transport of G-E2TMCT+ to the Golgi was rapid (t1/2 = 10-20 min). The G-E2TMCT+ protein was determined to be distal to or within the medial Golgi based on acquisition of endo H resistance but proximal to the trans-Golgi network since it lacked sialic acid. Deletion analysis revealed that only the TM domain of E2 was required for Golgi targeting. Although the cytoplasmic domain of E2 was not necessary for Golgi retention, it was required for efficient transport of VSV G-RV chimeras out of the endoplasmic reticulum. When assayed in sucrose velocity sedimentations gradients, the Golgi-retained G-E2TMCT+ protein behaved as a dimer. Unlike virtually all other Golgi targeting signals, the E2 TM domain does not contain any polar amino acids. The TM and CT domains of E1 were not required for targeting of E2 and E1 to the Golgi indicating that a heterodimer of two integral membrane proteins can be retained in the Golgi by a single retention signal.


Asunto(s)
Aparato de Golgi/metabolismo , Glicoproteínas de Membrana/metabolismo , Señales de Clasificación de Proteína/metabolismo , Virus de la Rubéola/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Transporte Biológico , Células CHO , Cricetinae , Membranas Intracelulares/metabolismo , Sustancias Macromoleculares , Glicoproteínas de Membrana/química , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Multimerización de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Virus de la Rubéola/genética , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
17.
Virology ; 202(2): 574-85, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8030223

RESUMEN

We have developed a stably transfected CHO cell line (CHO24S) that expresses the three structural proteins of rubella virus (RV). RV proteins C (capsid), E2, and E1 are secreted from CHO24S cells in the form of RV-like particles (RLPs) which form by budding into the cisterna of the Golgi complex. RLPs resemble RV virions in their size and morphology and have an identical buoyant density when purified on sucrose gradients. Release of RLPs into the medium was found to be dependent upon the E1 cytoplasmic tail since deletion or substitution of this domain with the same region from vesicular stomatitis virus G protein abrogated release of RV proteins from transfected cells. These results indicate that the RV 40S genomic RNA is not required for efficient particle assembly. Therefore, RLPs may serve as a convenient source of RV antigen for use in diagnostic assays and as an alternative to live attenuated vaccine strains.


Asunto(s)
Virus de la Rubéola/ultraestructura , Proteínas Virales , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/genética , Antígenos Virales/metabolismo , Secuencia de Bases , Células CHO , Cápside/ultraestructura , Cricetinae , Citoplasma/metabolismo , Cartilla de ADN/química , Aparato de Golgi/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Microscopía Electrónica , Datos de Secuencia Molecular , Morfogénesis , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes , Virus de la Rubéola/inmunología , Relación Estructura-Actividad , Transfección , Vacunas Sintéticas , Proteínas Virales/inmunología , Virión/ultraestructura
18.
J Virol ; 68(6): 4086-91, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8189549

RESUMEN

Rubella virus (RV) virions contain two envelope glycoproteins (E1 and E2) and a capsid protein (C). Noninfectious RV-like particles (VLPs) containing three structural proteins were expressed in a BHK cell line (BHK-24S) by using an inducible promoter. These VLPs were found to resemble RV virons in terms of their size, their morphology, and some biological activities. In immunoblotting studies, VLPs were found to bind similarly to native RV virions with 10 of a panel of 12 RV-specific murine monoclonal antibodies. Immunization of mice with VLPs induced specific antibody responses against RV structural proteins as well as virus-neutralizing and hemagglutination-inhibiting antibodies. After immunization of mice with VLPs, in vitro challenge of isolated lymphocytes with inactivated RV and individual RV structural proteins stimulated proliferation. Our data suggest the possibility of using VLPs as immunogens for serodiagnostic assays and RV vaccines.


Asunto(s)
Virus de la Rubéola/metabolismo , Virus de la Rubéola/ultraestructura , Proteínas Estructurales Virales/metabolismo , Animales , Anticuerpos Antivirales/biosíntesis , Antígenos Virales , Transformación Celular Viral , Cricetinae , ADN Viral/genética , Inmunización , Cuerpos de Inclusión Viral/inmunología , Cuerpos de Inclusión Viral/metabolismo , Cuerpos de Inclusión Viral/ultraestructura , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Virus de la Rubéola/genética , Virus de la Rubéola/inmunología , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/inmunología
19.
Virology ; 200(2): 821-5, 1994 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-8178466

RESUMEN

Endoproteolytic cleavage of precursors is a key step in biosynthesis of functional proteins. The structural proteins of rubella virus are initially translated as a precursor polyprotein in the order NH2-C-E2-E1-COOH and are cleaved by host signal peptidase to yield three structural proteins. Between regions corresponding to E2 and E1 in the precursor is a region of seven amino acid residues (R-R-A-C-R-R-R) that contains a motif for stop-transfer or a possible target for trypsin-like protease cleavage. Using site-directed mutagenesis, these arginine residues, as well as the signal peptide cleavage site at the N-terminus of E1, have been mutated individually or in combination. Results from in vitro transcription/translation analysis indicated that the mutated E2E1 precursor polyproteins were translocated into the microsome and glycosylated. Expression of mutated precursor polyproteins in COS cells revealed that the cleavage of E2E1 polyprotein precursor was impaired when the signal peptide cleavage site alone or both arginine clusters were altered, whereas partial cleavage was observed in the mutants in which either one of the two arginine clusters was modified. Our data suggest that although the arginine clusters do not function as a basic protease cleavage site, they contribute to maintain the proper configuration of that region for access of cellular signal peptidase.


Asunto(s)
Arginina/genética , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Virus de la Rubéola/genética , Proteínas del Envoltorio Viral/biosíntesis , Secuencia de Aminoácidos , Secuencia de Bases , Análisis Mutacional de ADN , Datos de Secuencia Molecular , Biosíntesis de Proteínas , Precursores de Proteínas/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Proteínas del Envoltorio Viral/genética
20.
Virus Res ; 31(3): 277-89, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8191784

RESUMEN

Rubella virus (RV) virions contain two envelope glycoproteins, E1 and E2. Removal of hydrophobic regions in their carboxyl termini by genetic engineering caused them to be secreted rather than maintained in cell membranes of transfected COS cells. Truncated E2 was secreted in the absence of E1, whereas E1 lacking its transmembrane domain required coexpression of E2 for export from the cell. Secreted E2 was found to contain both O-linked and N-linked complex glycans, whereas secreted E1 retained virus neutralization and hemagglutination epitopes, suggesting the possibility of using soluble RV antigens as subunit vaccines and for serodiagnostic purposes. Stable Chinese hamster ovary cell lines secreting RV E1 were constructed for large scale preparation of recombinant E1.


Asunto(s)
Proteínas del Envoltorio Viral/biosíntesis , Animales , Secuencia de Bases , Células CHO/metabolismo , Células Cultivadas , Cricetinae , ADN Viral/genética , Regulación Viral de la Expresión Génica , Genes Virales/genética , Riñón , Mamíferos , Datos de Secuencia Molecular , Transfección , Proteínas del Envoltorio Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...