Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 75(22): 11034-55, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11602744

RESUMO

In a series of papers, we have provided evidence that during its assembly vaccinia virus is enveloped by a membrane cisterna that originates from a specialized, virally modified, smooth-membraned domain of the endoplasmic reticulum (ER). Recently, however, Hollinshead et al. (M. Hollinshead, A. Vanderplasschen, G. I. Smith, and D. J. Vaux, J. Virol. 73:1503-1517, 1999) argued against this hypothesis, based on their interpretations of thin-sectioned material. The present article is the first in a series of papers that describe a comprehensive electron microscopy (EM) analysis of the vaccinia Intracellular Mature Virus (IMV) and the process of its assembly in HeLa cells. In this first study, we analyzed the IMV by on-grid staining, cryo-scanning EM (SEM), and cryo-transmission EM. We focused on the structure of the IMV particle, both after isolation and in the context of viral entry. For the latter, we used high-resolution cryo-SEM combined with cryofixation, as well as a novel approach we developed for investigating vaccinia IMV bound to plasma membrane fragments adsorbed onto EM grids. Our analysis revealed that the IMV is made up of interconnected cisternal and tubular domains that fold upon themselves via a complex topology that includes an S-shaped fold. The viral tubules appear to be eviscerated from the particle during viral infection. Since the structure of the IMV is the result of a complex assembly process, we also provide a working model to explain how a specialized smooth-ER domain can be modulated to form the IMV. We also present theoretical arguments for why it is highly unlikely that the IMV is surrounded by only a single membrane.


Assuntos
Vaccinia virus/ultraestrutura , Vírion/ultraestrutura , Montagem de Vírus , Ditiotreitol/farmacologia , Células HeLa , Humanos , Microscopia Eletrônica de Varredura , Vaccinia virus/fisiologia , Vírion/fisiologia
2.
J Virol ; 75(22): 11056-70, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11602745

RESUMO

In the preceding study (see accompanying paper), we showed by a variety of different techniques that intracellular mature vaccinia virus (vaccinia IMV) is unexpectedly complex in its structural organization and that this complexity also extends to the underlying viral core, which is highly folded. With that analysis as a foundation, we now present different thin-section electron microscopy approaches for analyzing the IMV and the processes by which it is assembled in infected HeLa cells. We focus on conventional epoxy resin thin sections as well as cryosections to describe key intermediates in the assembly process. We took advantage of streptolysin O's ability to selectively permeabilize the plasma membrane of infected cells to improve membrane contrast, and we used antibodies against bone fide integral membrane proteins of the virus to unequivocally identify membrane profiles in thin sections. All of the images presented here can be rationalized with respect to the model put forward for the assembly of the IMV in the accompanying paper.


Assuntos
Vaccinia virus/ultraestrutura , Montagem de Vírus , Proteínas de Bactérias , Crioultramicrotomia , DNA Viral/análise , Retículo Endoplasmático Rugoso/virologia , Células HeLa , Humanos , Microscopia Eletrônica , Estreptolisinas/farmacologia , Vaccinia virus/isolamento & purificação , Vaccinia virus/fisiologia , Proteínas do Envelope Viral/análise
3.
Mol Biol Cell ; 11(7): 2497-511, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10888684

RESUMO

The simpler of the two infectious forms of vaccinia virus, the intracellular mature virus (IMV) is known to infect cells less efficiently than the extracellular enveloped virus (EEV), which is surrounded by an additional, TGN-derived membrane. We show here that when the IMV binds HeLa cells, it activates a signaling cascade that is regulated by the GTPase rac1 and rhoA, ezrin, and both tyrosine and protein kinase C phosphorylation. These cascades are linked to the formation of actin and ezrin containing protrusions at the plasma membrane that seem to be essential for the entry of IMV cores. The identical cores of the EEV also appear to enter at the cell surface, but surprisingly, without the need for signaling and actin/membrane rearrangements. Thus, in addition to its known role in wrapping the IMV and the formation of intracellular actin comets, the membrane of the EEV seems to have evolved the capacity to enter cells silently, without a need for signaling.


Assuntos
Membrana Celular/virologia , Transdução de Sinais , Vaccinia virus/fisiologia , Actinas/metabolismo , Proteínas do Citoesqueleto , Células HeLa , Humanos , Fosfoproteínas/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Vaccinia virus/metabolismo , Vaccinia virus/patogenicidade , Vírion/metabolismo , Montagem de Vírus , Proteínas rho de Ligação ao GTP/metabolismo
4.
J Virol ; 74(8): 3525-36, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10729126

RESUMO

The entry of vaccinia virus (VV) into the host cell results in the delivery of the double-stranded DNA genome-containing core into the cytoplasm. The core is disassembled, releasing the viral DNA in order to initiate VV cytoplasmic transcription and DNA replication. Core disassembly can be prevented using the VV early transcription inhibitor actinomycin D (actD), since early VV protein synthesis is required for core uncoating. In this study, VV intracellular cores were accumulated in the presence of actD and isolated from infected cells. The content of these cores was analyzed by negative staining EM and by Western blotting using a collection of antibodies to VV core and membrane proteins. By Western blot analyses, intracellular actD cores, as well as cores prepared by NP-40-dithiothreitol treatment of purified virions (NP-40/DTT cores), contained the core proteins p25 (encoded by L4R), 4a (A10L), 4b (A3L), and p39 (A4L) as well as small amounts of the VV membrane proteins p32 (D8L) and p35 (H3L). While NP-40/DTT cores contained the major putative DNA-binding protein p11 (F17R), actD cores entirely lacked this protein. Labeled cryosections of cells infected for different periods of time in the presence or absence of actD were subsequently used to follow the fate of VV core proteins by EM. These EM images confirmed that p11 was lost at the plasma membrane upon core penetration. The cores that accumulated in the presence of actD were labeled with antibodies to 4a, p39, p25, and DNA at all times examined. In the absence of the drug the cores gradually lost their electron-dense inner part, concomitant with the loss of p25 and DNA labeling. The remaining core shell still labeled with antibodies to p39 and 4a/4b, implying that these proteins are part of this structure. These combined data are discussed with respect to the structure of VV as well as core disassembly.


Assuntos
Vaccinia virus/metabolismo , Vaccinia virus/ultraestrutura , Proteínas do Core Viral/metabolismo , Western Blotting , Ditiotreitol/farmacologia , Ensaio de Imunoadsorção Enzimática , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Microscopia Eletrônica , Octoxinol , Polietilenoglicóis/farmacologia , Vaccinia virus/crescimento & desenvolvimento , Vaccinia virus/patogenicidade , Proteínas do Core Viral/ultraestrutura , Proteínas Virais/metabolismo , Proteínas Virais/ultraestrutura , Montagem de Vírus
5.
J Virol ; 74(5): 2333-42, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10666264

RESUMO

The small envelope (E) protein has recently been shown to play an essential role in the assembly of coronaviruses. Expression studies revealed that for formation of the viral envelope, actually only the E protein and the membrane (M) protein are required. Since little is known about this generally low-abundance virion component, we have characterized the E protein of mouse hepatitis virus strain A59 (MHV-A59), an 83-residue polypeptide. Using an antiserum to the hydrophilic carboxy terminus of this otherwise hydrophobic protein, we found that the E protein was synthesized in infected cells with similar kinetics as the other viral structural proteins. The protein appeared to be quite stable both during infection and when expressed individually using a vaccinia virus expression system. Consistent with the lack of a predicted cleavage site, the protein was found to become integrated in membranes without involvement of a cleaved signal peptide, nor were any other modifications of the polypeptide observed. Immunofluorescence analysis of cells expressing the E protein demonstrated that the hydrophilic tail is exposed on the cytoplasmic side. Accordingly, this domain of the protein could not be detected on the outside of virions but appeared to be inside, where it was protected from proteolytic degradation. The results lead to a topological model in which the polypeptide is buried within the membrane, spanning the lipid bilayer once, possibly twice, and exposing only its carboxy-terminal domain. Finally, electron microscopic studies demonstrated that expression of the E protein in cells induced the formation of characteristic membrane structures also observed in MHV-A59-infected cells, apparently consisting of masses of tubular, smooth, convoluted membranes. As judged by their colabeling with antibodies to E and to Rab-1, a marker for the intermediate compartment and endoplasmic reticulum, the E protein accumulates in and induces curvature into these pre-Golgi membranes where coronaviruses have been shown earlier to assemble by budding.


Assuntos
Coronavirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Membrana Celular/virologia , Coronavirus/química , Imunofluorescência , Vetores Genéticos , Células L , Camundongos , Microscopia Eletrônica , Vírus da Hepatite Murina/metabolismo , Testes de Precipitina , Proteínas Recombinantes/metabolismo , Transfecção , Vaccinia virus/genética , Proteínas do Envelope Viral/análise , Proteínas do Envelope Viral/genética , Montagem de Vírus , Integração Viral
6.
J Virol ; 73(9): 7641-57, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10438855

RESUMO

The aim of the present study was to define the site of replication of the coronavirus mouse hepatitis virus (MHV). Antibodies directed against several proteins derived from the gene 1 polyprotein, including the 3C-like protease (3CLpro), the putative polymerase (POL), helicase, and a recently described protein (p22) derived from the C terminus of the open reading frame 1a protein (CT1a), were used to probe MHV-infected cells by indirect immunofluorescence (IF) and electron microscopy (EM). At early times of infection, all of these proteins showed a distinct punctate labeling by IF. Antibodies to the nucleocapsid protein also displayed a punctate labeling that largely colocalized with the replicase proteins. When infected cells were metabolically labeled with 5-bromouridine 5'-triphosphate (BrUTP), the site of viral RNA synthesis was shown by IF to colocalize with CT1a and the 3CLpro. As shown by EM, CT1a localized to LAMP-1 positive late endosomes/lysosomes while POL accumulated predominantly in multilayered structures with the appearance of endocytic carrier vesicles. These latter structures were also labeled to some extent with both anti-CT1a and LAMP-1 antibodies and could be filled with fluid phase endocytic tracers. When EM was used to determine sites of BrUTP incorporation into viral RNA at early times of infection, the viral RNA localized to late endosomal membranes as well. These results demonstrate that MHV replication occurs on late endosomal membranes and that several nonstructural proteins derived from the gene 1 polyprotein may participate in the formation and function of the viral replication complexes.


Assuntos
Vírus da Hepatite Murina/química , RNA Viral/biossíntese , Proteínas não Estruturais Virais/análise , Proteínas Virais/análise , Replicação Viral , Animais , Anticorpos Antivirais/imunologia , Antígenos CD/análise , Endocitose , Endossomos , Técnica Indireta de Fluorescência para Anticorpo , Células L , Proteínas de Membrana Lisossomal , Glicoproteínas de Membrana/análise , Camundongos , Microscopia de Fluorescência , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/fisiologia , Proteínas do Nucleocapsídeo/análise , Fases de Leitura Aberta , RNA Helicases/análise , RNA Polimerase Dependente de RNA/análise , Frações Subcelulares
7.
J Cell Biol ; 144(2): 267-79, 1999 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-9922453

RESUMO

Previous data have shown that reducing agents disrupt the structure of vaccinia virus (vv). Here, we have analyzed the disulfide bonding of vv proteins in detail. In vv-infected cells cytoplasmically synthesized vv core proteins became disulfide bonded in the newly assembled intracellular mature viruses (IMVs). vv membrane proteins also assembled disulfide bonds, but independent of IMV formation and to a large extent on their cytoplasmic domains. If disulfide bonding was prevented, virus assembly was only partially impaired as shown by electron microscopy as well as a biochemical assay of IMV formation. Under these conditions, however, the membranes around the isolated particles appeared less stable and detached from the underlying core. During the viral infection process the membrane proteins remained disulfide bonded, whereas the core proteins were reduced, concomitant with delivery of the cores into the cytoplasm. Our data show that vv has evolved an unique system for the assembly of cytoplasmic disulfide bonds that are localized both on the exterior and interior parts of the IMV.


Assuntos
Dissulfetos/metabolismo , Vaccinia virus/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas do Envelope Viral/metabolismo , Antivirais/farmacologia , Cisteína/metabolismo , Citoplasma , Citosol/metabolismo , Dimerização , Ditiotreitol/farmacologia , Etilmaleimida/metabolismo , Etilmaleimida/farmacologia , Células HeLa , Humanos , Cinética , Reagentes de Sulfidrila/metabolismo , Reagentes de Sulfidrila/farmacologia , Trítio , Vaccinia virus/efeitos dos fármacos
8.
J Virol ; 72(8): 6689-98, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9658116

RESUMO

Among the functions of the replicase of equine arteritis virus (EAV; family Arteriviridae, order Nidovirales) are important viral enzyme activities such as proteases and the putative RNA polymerase and RNA helicase functions. The replicase is expressed in the form of two polyproteins (open reading frame 1a [ORF1a] and ORF1ab), which are processed into 12 nonstructural proteins by three viral proteases. In immunofluorescence assays, the majority of these cleavage products localized to the perinuclear region of the cell. A dense granular and vesicular staining was observed, which strongly suggested membrane association. By using confocal microscopy and double-label immunofluorescence, the distribution of the EAV replicase was shown to overlap with that of PDI, a resident protein of the endoplasmic reticulum and intermediate compartment. An in situ labeling of nascent viral RNA with bromo-UTP demonstrated that the membrane-bound complex in which the replicase subunits accumulate is indeed the site of viral RNA synthesis. A number of ORF1a-encoded hydrophobic domains were postulated to be involved in the membrane association of the arterivirus replication complex. By using various biochemical methods (Triton X-114 extraction, membrane purification, and sodium carbonate treatment), replicase subunits containing these domains were shown to behave as integral membrane proteins and to be membrane associated in infected cells. Thus, contribution to the formation of a membrane-bound scaffold for the viral replication-transcription complex appears to be an important novel function for the arterivirus ORF1a replicase polyprotein.


Assuntos
Equartevirus/enzimologia , Fases de Leitura Aberta , RNA Polimerase Dependente de RNA/metabolismo , Replicação Viral , Animais , Linhagem Celular , Membrana Celular/enzimologia , Membrana Celular/virologia , Núcleo Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Retículo Endoplasmático/metabolismo , Equartevirus/genética , Equartevirus/fisiologia , Octoxinol , Polietilenoglicóis , RNA Viral/biossíntese , Coelhos , Células Vero
9.
J Virol ; 71(10): 7404-20, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9311819

RESUMO

The use of two-dimensional gel electrophoresis has identified the gene products A14L (p16) and A13L (p8) as abundant membrane proteins of the first infectious form of vaccinia virus, the intracellular mature virus (IMV; O. N. Jensen, T. Houthaeve, A. Shevchenko, S. Cudmore, T. Ashford, M. Mann, G. Griffiths, J. Krijnse Locker, J. Virol. 70:7485-7497, 1996). In this study, these two proteins were characterized in detail. In infected cells, both proteins localize not only to the viral membranes but also to tubular-cisternal membranes of the intermediate compartment, defined by the use of antibodies to either rab1A or p21, which colocalize with rab1A (J. Krijnse Locker, S. Schleich, D. Rodriguez, B. Goud, E. J. Snijder, and G. Griffiths, J. Biol. Chem. 271:14950-14958, 1996). Both proteins appear to reach this destination via cotranslational insertion into the rough endoplasmic reticulum, as shown by in vitro translation and translocation experiments. Whereas p16 probably spans the membrane twice, p8 is inserted into the membrane by means of its single NH2-terminal hydrophobic domain, adopting a topology which leaves the C terminus exposed to the cytoplasm. Combined immunocytochemical and biochemical data show that p16 is a member of the inner of the two IMV membrane layers, whereas p8 localizes to both the inner and the outer membrane. These findings are discussed with respect to our model of IMV membrane assembly.


Assuntos
Retículo Endoplasmático Rugoso/fisiologia , Biossíntese de Proteínas , Vaccinia virus/fisiologia , Proteínas Virais/biossíntese , Western Blotting , Proteínas de Ligação ao Cálcio/análise , Calnexina , Permeabilidade da Membrana Celular , Eletroforese em Gel Bidimensional , Retículo Endoplasmático Rugoso/ultraestrutura , Retículo Endoplasmático Rugoso/virologia , Técnica Indireta de Fluorescência para Anticorpo , Células HeLa , Humanos , Microscopia Imunoeletrônica , Processamento de Proteína Pós-Traducional , Vaccinia virus/ultraestrutura , Proteínas Virais/análise
10.
Virology ; 235(2): 218-27, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9281501

RESUMO

A novel method is described which facilitates the in vitro assembly of one step in the life cycle of vaccinia virus, the formation of the spherical immature virus (IV). For this, advantage was taken of the ability of rifampicin to reversibly block the assembly of the IV. Rifampicin-treated, vaccinia virus-infected HeLa cells were permeabilized with streptolysin O (SLO) and the endogenous cytosol was allowed to exit the cells at 4 degrees . Subsequently, exogenous cytosol from infected or uninfected HeLa cells as well as an ATP-regenerating system were added and the cells were incubated for different times at 37 degrees in the absence of rifampicin. The preparations were then evaluated by thin section EM. Our data show that in the presence of infected or uninfected cell cytosol and ATP a significant fraction of cells could reconstitute IV assembly in vitro. Under no conditions were we able to reconstitute any later stages of assembly. The potential of this system for the in vitro reconstitution of viral assembly in general is discussed.


Assuntos
Rifampina/farmacologia , Estreptolisinas/farmacologia , Vaccinia virus/fisiologia , Trifosfato de Adenosina/fisiologia , Proteínas de Bactérias , Citosol/fisiologia , Células HeLa/química , Células HeLa/ultraestrutura , Humanos , Imuno-Histoquímica , Proteínas de Membrana/análise , Microscopia Eletrônica , Vaccinia virus/química , Proteínas do Envelope Viral/análise , Proteínas Virais/análise , Montagem de Vírus
11.
J Virol ; 69(11): 7072-86, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7474127

RESUMO

We have characterized a temperature-sensitive mutant of vaccinia virus, ts16, originally isolated by Condit et al. (Virology 128:429-443, 1983), at the permissive and nonpermissive temperatures. In a previous study by Kane and Shuman (J. Virol 67:2689-2698, 1993), the mutation of ts16 was mapped to the I7 gene, encoding a 47-kDa protein that shows partial homology to the type II topoisomerase of Saccharomyces cerevisiae. The present study extends previous electron microscopy analysis, showing that in BSC40 cells infected with ts16 at the restrictive temperature (40 degrees C), the assembly was arrested at a stage between the spherical immature virus and the intracellular mature virus (IMV). In thawed cryosections, a number of the major proteins normally found in the IMV were subsequently localized to these mutant particles. By using sucrose density gradients, the ts16 particles were purified from cells infected at the permissive and nonpermissive temperatures. These were analyzed by immunogold labelling and negative-staining electron microscopy, and their protein composition was determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. While the ts16 virus particles made at the permissive temperature appeared to have a protein pattern identical to that of wild-type IMV, in the mutant particles the three core proteins, p4a, p4b, and 28K, were not proteolytically processed. Consistent with previous data the sucrose-purified particles could be labelled with [3H]thymidine. In addition, anti-DNA labelling on thawed cryosections suggested that most of the mutant particles had taken up DNA. On thawed cryosections of cells infected at the permissive temperature, antibodies to I7 labelled the virus factories, the immature viruses, and the IMVs, while under restrictive conditions these structures were labelled much less, if at all. Surprisingly, however, by Western blotting (immunoblotting) the I7 protein was present in similar amounts in the defective particles and in the IMVs isolated at the permissive temperature. Finally, our data suggest that at the nonpermissive temperature the assembly of ts16 is irreversibly arrested in a stage at which the DNA is in the process of entering but before the particle has completely sealed, as monitored by protease experiments.


Assuntos
Genes Virais , Mutação , Vaccinia virus/genética , Vaccinia virus/ultraestrutura , Proteínas Virais/análise , Animais , Linhagem Celular , DNA Topoisomerases Tipo II/biossíntese , DNA Topoisomerases Tipo II/genética , DNA Viral/análise , DNA Viral/biossíntese , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Vírus Defeituosos/ultraestrutura , Resinas Epóxi , Células HeLa , Humanos , Microscopia Eletrônica , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Temperatura , Vaccinia virus/fisiologia , Proteínas Virais/biossíntese
12.
J Cell Sci ; 108 ( Pt 8): 2839-56, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7593324

RESUMO

Recent data strongly suggest that the coatomer (COP) complex is involved in membrane transport between the ER and Golgi complex. This vesicular coat has been implicated in ER to Golgi, in intra Golgi as well as in Golgi to ER traffic. In this study we present a detailed immunocytochemical analysis of the distribution of beta-COP in different tissue culture cells. Our results extend previous studies by showing, using electron microscopy, that beta-COP accumulates on vesicular profiles and buds in the intermediate compartment (IC) under conditions that block ER to Golgi transport (15 degrees C). Importantly, under these conditions beta-COP co-localizes on these structures with a passenger protein, the membrane glycoprotein of vesicular stomatis virus (ts-O45-G). Furthermore, quantitative immunofluorescence microscopy of cells with ts-045-G accumulated in the ER, IC and trans-Golgi network, shifted briefly to the permissive temperature, showed that beta-COP was associated with many of the putative transport intermediates containing the viral glycoprotein which is in transit between the ER/IC and the cis-Golgi. The simplest interpretation of these data is that COP-coated vesicles are involved in anterograde transport of ts-045-G from the IC to the Golgi complex. Since many putative COP vesicle lacked the G protein following release of the 15 degrees C block this pool could be involved in retrograde transport. We also show that beta-COP is present on the membranes of the trans-Golgi network. However, in contrast to the ER-Golgi boundary, we could find no convincing evidence that this pool of beta-COP is associated with buds or trans-Golgi network-derived transport vesicles.


Assuntos
Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/ultraestrutura , Proteínas Associadas aos Microtúbulos/análise , Animais , Chlorocebus aethiops , Proteína Coatomer , Cicloeximida/farmacologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Proteínas de Ligação ao GTP/análise , Proteínas de Ligação ao GTP/biossíntese , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica/métodos , Células L , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Camundongos , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Sialiltransferases/análise , Sialiltransferases/biossíntese , Células Vero , Vírus da Estomatite Vesicular Indiana
13.
J Biol Chem ; 270(15): 8815-21, 1995 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-7721788

RESUMO

The mouse hepatitis virus M protein is a triple spanning membrane glycoprotein that, when expressed independently, localizes to trans-Golgi as well as to the trans-Golgi network (TGN). Passage of this protein from the endoplasmic reticulum through the intermediate compartment to the late Golgi and TGN can be conveniently followed by analyzing its O-linked sugars. Using pulse-chase analyses we studied the oligomerization of the M protein in sucrose gradients. The Golgi and TGN forms migrated as large heterogeneous complexes, whereas the endoplasmic reticulum and intermediate compartment forms of the protein appeared to migrate as monomer. Moreover, a mutant of the M protein lacking the 22 COOH-terminal amino acids, that is transported to the plasma membrane, gave rise to similar complexes, albeit smaller in size, that persisted at the plasma membrane. We propose that the trans-Golgi/TGN retention of the MHV-M protein is governed by two mechanisms: oligomerization possibly mediated by the transmembrane domains and binding of its cytoplasmic tail to cellular factors in trans Golgi/TGN.


Assuntos
Complexo de Golgi/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Biopolímeros , Linhagem Celular , Proteínas M de Coronavírus , Complexo de Golgi/ultraestrutura , Microscopia Eletrônica , Vírus da Hepatite Murina/metabolismo , Proteínas da Matriz Viral/ultraestrutura
14.
J Biol Chem ; 269(45): 28263-9, 1994 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-7961764

RESUMO

The M protein of mouse hepatitis virus (MHV) is a triple-spanning membrane glycoprotein that is exclusively O-glycosylated. When expressed independently, it accumulates in late Golgi and the trans-Golgi network (TGN) (Locker, J. K., Griffiths, G., Horzinek, M. C., and Rottier, P. J. M. (1992) (J. Biol. Chem. 267, 14094-14101). To analyze the domains of this protein responsible for its localization, we have generated deletion mutants by site-directed mutagenesis and analyzed their intracellular transport. The intracellular distribution of the mutant proteins was determined by following the extent of O-glycosylation in pulse-chase experiments, by electron microscopic immunocytochemistry, and by surface immunoprecipitation. Mutant proteins lacking the first or the first and second transmembrane domains were not efficiently retained in the Golgi complex or TGN. The latter mutant proteins also localized to endocytic compartments but were not subject to rapid lysosomal degradation. Deletion of the COOH-terminal 22 amino acids, including a tyrosine residue in the context of a potential internalization signal, resulted in plasma membrane exposure of the respective mutant protein. We show that the wild-type MHV-M protein does not recycle between the plasma membrane and the TGN, but rather behaves as a late Golgi/TGN resident in our assays. We propose that the MHV-M protein is retained in the Golgi by two signals, one contained in the cytoplasmic tail and the other determined by the transmembrane domains.


Assuntos
Complexo de Golgi/metabolismo , Vírus da Hepatite Murina/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Sequência de Carboidratos , Carcinoma Hepatocelular , Linhagem Celular , Chlorocebus aethiops , Endossomos/metabolismo , Glicosilação , Humanos , Rim , Neoplasias Hepáticas , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Vírus da Hepatite Murina/genética , Deleção de Sequência , Transfecção , Proteínas da Matriz Viral/biossíntese , Proteínas da Matriz Viral/química
15.
J Virol ; 68(10): 6523-34, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8083990

RESUMO

The prevailing hypothesis is that the intracellular site of budding of coronaviruses is determined by the localization of its membrane protein M (previously called E1). We tested this by analyzing the site of budding of four different coronaviruses in relation to the intracellular localization of their M proteins. Mouse hepatitis virus (MHV) and infectious bronchitis virus (IBV) grown in Sac(-) cells, and feline infectious peritonitis virus (FIPV) and transmissible gastroenteritis virus (TGEV) grown in CrFK cells, all budded exclusively into smooth-walled, tubulovesicular membranes located intermediately between the rough endoplasmic reticulum and Golgi complex, identical to the so-called budding compartment previously identified for MHV. Indirect immunofluorescence staining of the infected cells showed that all four M proteins accumulated in a perinuclear region. Immunogold microscopy localized MHV M and IBV M in the budding compartment; in addition, a dense labeling in the Golgi complex occurred, MHV M predominantly in trans-Golgi cisternae and trans-Golgi reticulum and IBV M mainly in the cis and medial Golgi cisternae. The corresponding M proteins of the four viruses, when independently expressed in a recombinant vaccinia virus system, also accumulated in the perinuclear area. Quantitative pulse-chase analysis of metabolically labeled cells showed that in each case the majority of the M glycoproteins carried oligosaccharide side chains with Golgi-specific modifications within 4 h after synthesis. Immunoelectron microscopy localized recombinant MHV M and IBV M to the same membranes as the respective proteins in coronavirus-infected cells, with the same cis-trans distribution over the Golgi complex. Our results demonstrate that some of the M proteins of the four viruses are transported beyond the budding compartment and are differentially retained by intrinsic retention signals; in addition to M, other viral and/or cellular factors are probably required to determine the site of budding.


Assuntos
Coronaviridae/crescimento & desenvolvimento , Complexo de Golgi/microbiologia , Proteínas da Matriz Viral/biossíntese , Animais , Gatos , Linhagem Celular , Coronaviridae/metabolismo , Coronaviridae/ultraestrutura , Coronavirus Felino/crescimento & desenvolvimento , Coronavirus Felino/metabolismo , Coronavirus Felino/ultraestrutura , Feto , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Humanos , Vírus da Bronquite Infecciosa/crescimento & desenvolvimento , Vírus da Bronquite Infecciosa/metabolismo , Vírus da Bronquite Infecciosa/ultraestrutura , Rim , Microscopia Eletrônica , Microscopia Imunoeletrônica , Vírus da Hepatite Murina/crescimento & desenvolvimento , Vírus da Hepatite Murina/metabolismo , Vírus da Hepatite Murina/ultraestrutura , Recombinação Genética , Suínos , Vírus da Gastroenterite Transmissível/crescimento & desenvolvimento , Vírus da Gastroenterite Transmissível/metabolismo , Vírus da Gastroenterite Transmissível/ultraestrutura , Células Tumorais Cultivadas , Proteínas da Matriz Viral/análise
16.
J Biol Chem ; 267(30): 21911-8, 1992 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-1400501

RESUMO

The M protein of mouse hepatitis virus strain A59 is a triple-spanning membrane protein which assembles with an uncleaved internal signal sequence, adopting an NexoCcyt orientation. To study the insertion mechanism of this protein, domains potentially involved in topogenesis were deleted and the effects analyzed in topogenesis were deleted and the effects analyzed in several ways. Mutant proteins were synthesized in a cell-free translation system in the presence of microsomal membranes, and their integration and topology were determined by alkaline extraction and by protease-protection experiments. By expression in COS-1 and Madin-Darby canine kidney-II cells, the topology of the mutant proteins was also analyzed in vivo. Glycosylation was used as a biochemical marker to assess the disposition of the NH2 terminus. An indirect immunofluorescence assay on semi-intact Madin-Darby canine kidney-II cells using domain-specific antibodies served to identify the cytoplasmically exposed domains. The results show that each membrane-spanning domain acts independently as an insertion and anchor signal and adopts an intrinsic preferred orientation in the lipid bilayer which corresponds to the disposition of the transmembrane domain in the wild-type assembled protein. These observations provide further insight into the mechanism of membrane integration of multispanning proteins. A model for the insertion of the coronavirus M protein is proposed.


Assuntos
Coronaviridae/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas da Matriz Viral/metabolismo , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Coronaviridae/genética , Cães , Imunofluorescência , Glicosilação , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação , Conformação Proteica , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética
17.
J Biol Chem ; 267(20): 14094-101, 1992 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-1629209

RESUMO

It has previously been shown that the M (E1) glycoprotein of mouse hepatitis virus strain A59 (MHV-A59) contains only O-linked oligosaccharides and localizes to the Golgi region when expressed independently. A detailed pulse-chase analysis was made of the addition of O-linked sugars to the M protein; upon sodium dodecyl sulfate-polyacrylamide gel electrophoresis, three different electrophoretic forms could be distinguished that corresponded to the sequential acquisition of N-acetylgalactosamine (GalNAc), galactose (Gal), and sialic acid (SA). A fourth and fifth form could also be detected which we were unable to identify. Following Brefeldin A treatment, the M protein still acquired GalNAc, Gal, and SA, but the fourth and fifth forms were absent, suggesting that these modifications occur in the trans-Golgi network (TGN). In contrast, in the presence of BFA, the G protein of vesicular stomatitis virus (VSV), which contains N-linked oligosaccharides, acquired Gal and fucose but not SA. These results are consistent with earlier published data showing that Golgi compartments proximal to the TGN, but not the TGN itself, relocate to the endoplasmatic reticulum/intermediate compartment. More importantly, our data argue that, whereas addition of SA to N-linked sugars occurs in the TGN the acquisition of both SA on O-linked sugars and the addition of fucose to N-linked oligosaccharides must occur in Golgi compartments proximal to the TGN. The glycosylation of the M protein moreover indicates that it is transported to trans-Golgi and TGN. This was confirmed by electron microscopy immunocytochemistry, showing that the protein is targeted to cisternae on the trans side of the Golgi and co-localizes, at least in part, with TGN 38, a marker of the TGN, as well as with a lectin specific for sialic acid.


Assuntos
Coronaviridae/metabolismo , Oligossacarídeos/biossíntese , Ácidos Siálicos/metabolismo , Sialiltransferases/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Antivirais/farmacologia , Brefeldina A , Linhagem Celular , Coronaviridae/efeitos dos fármacos , Coronaviridae/ultraestrutura , Ciclopentanos/farmacologia , Fucose/metabolismo , Galactose/metabolismo , Glucosamina/metabolismo , Glicosilação , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Cinética , Metionina/metabolismo , Ácido N-Acetilneuramínico , Neuraminidase , Oligossacarídeos/isolamento & purificação , Radioisótopos de Enxofre , Trítio , Proteínas da Matriz Viral/biossíntese , Proteínas da Matriz Viral/isolamento & purificação
18.
Virology ; 182(2): 655-63, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2024492

RESUMO

The nucleotide sequence of the Berne virus envelope (E) protein gene was determined and its 26.5K translation product was identified by in vitro transcription and translation. Computer analysis of the protein sequence revealed the characteristics of a class III membrane protein lacking a cleaved signal sequence but containing three successive transmembrane alpha-helices in the N-terminal half, much the same as the coronavirus membrane (M) protein. The disposition of the E protein in the membrane was studied by in vitro translation in the presence of microsomes and by subsequent proteinase K digestion. Only small portions of either end of the polypeptide were found to be exposed on opposite sides of the vesicle membranes. Experiments with a hybrid E protein (EM) containing the C-terminal tail of a coronavirus M protein, to which an anti-peptide serum was available, showed that this C-terminus was present at the cytoplasmic side of the membrane, which is another similarity to the coronavirus M protein. Immunofluorescence experiments indicated that the EM protein, expressed by a recombinant vaccinia virus, accumulated in intracellular membranes, predominantly those of the endoplasmic reticulum. The common features of the torovirus E and the coronavirus M protein support our hypothesis that an evolutionary relationship exists between these groups of intracellularly budding viruses.


Assuntos
Vírus de RNA/genética , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Compartimento Celular , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , DNA/genética , Genes Virais , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Dados de Sequência Molecular , Peso Molecular , Vírus de RNA/análise , RNA Viral/genética , Termodinâmica , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/ultraestrutura , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...