Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Mol Biol ; 433(10): 166923, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33713677

RESUMO

How retroviral Gag proteins recognize the packaging signals (Psi) on their genomic RNA (gRNA) is a key question that we addressed here using Mason-Pfizer monkey virus (MPMV) as a model system by combining band-shift assays and footprinting experiments. Our data show that Pr78Gag selects gRNA against spliced viral RNA by simultaneously binding to two single stranded loops on the MPMV Psi RNA: (1) a large purine loop (ssPurines), and (2) a loop which partially overlaps with a mostly base-paired purine repeat (bpPurines) and extends into a GU-rich binding motif. Importantly, this second Gag binding site is located immediately downstream of the major splice donor (mSD) and is thus absent from the spliced viral RNAs. Identifying elements crucial for MPMV gRNA packaging should help in understanding not only the mechanism of virion assembly by retroviruses, but also facilitate construction of safer retroviral vectors for human gene therapy.


Assuntos
Produtos do Gene gag/química , Guanina/química , Vírus dos Macacos de Mason-Pfizer/química , RNA Viral/química , Uracila/química , Animais , Pareamento de Bases , Sequência de Bases , Sítios de Ligação , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Viral da Expressão Gênica , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Guanina/metabolismo , Interações Hospedeiro-Patógeno , Vírus dos Macacos de Mason-Pfizer/genética , Vírus dos Macacos de Mason-Pfizer/metabolismo , Conformação de Ácido Nucleico , Papio , Ligação Proteica , Conformação Proteica , Pegadas de Proteínas , RNA Viral/genética , RNA Viral/metabolismo , Transdução de Sinais , Uracila/metabolismo
2.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795446

RESUMO

Embryonic carcinoma (EC) cells are malignant counterparts of embryonic stem (ES) cells and serve as useful models for investigating cellular differentiation and human embryogenesis. Though the susceptibility of murine EC cells to retroviral infection has been extensively analyzed, few studies of retrovirus infection of human EC cells have been performed. We tested the susceptibility of human EC cells to transduction by retroviral vectors derived from three different retroviral genera. We show that human EC cells efficiently express reporter genes delivered by vectors based on human immunodeficiency virus type 1 (HIV-1) and Mason-Pfizer monkey virus (M-PMV) but not Moloney murine leukemia virus (MLV). In human EC cells, MLV integration occurs normally, but no viral gene expression is observed. The block to MLV expression of MLV genomes is relieved upon cellular differentiation. The lack of gene expression is correlated with transcriptional silencing of the MLV promoter through the deposition of repressive histone marks as well as DNA methylation. Moreover, depletion of SETDB1, a histone methyltransferase, resulted in a loss of transcriptional silencing and upregulation of MLV gene expression. Finally, we provide evidence showing that the lack of MLV gene expression may be attributed in part to the lack of MLV enhancer function in human EC cells. IMPORTANCE: Human embryonic carcinoma (EC) cells are shown to restrict the expression of murine leukemia virus genomes but not retroviral genomes of the lentiviral or betaretroviral families. The block occurs at the level of transcription and is accompanied by the deposition of repressive histone marks and methylation of the integrated proviral DNA. The host machinery required for silencing in human EC cells is distinct from that in murine EC cell lines: the histone methyltransferase SETDB1 is required, but the widely utilized corepressor TRIM28/Kap1 is not. A transcriptional enhancer element from the Mason-Pfizer monkey virus can override the silencing and promote transcription of chimeric proviral DNAs. The findings reveal novel features of human EC gene regulation not present in their murine counterparts.


Assuntos
Inativação Gênica , Genoma Viral , HIV-1/genética , Células-Tronco Embrionárias Humanas/imunologia , Vírus dos Macacos de Mason-Pfizer/genética , Vírus da Leucemia Murina de Moloney/genética , Células-Tronco Neoplásicas/imunologia , Animais , Diferenciação Celular , Metilação de DNA , Genes Reporter , HIV-1/metabolismo , Histona-Lisina N-Metiltransferase , Histonas/genética , Histonas/imunologia , Especificidade de Hospedeiro , Células-Tronco Embrionárias Humanas/virologia , Humanos , Vírus dos Macacos de Mason-Pfizer/metabolismo , Camundongos , Vírus da Leucemia Murina de Moloney/metabolismo , Células-Tronco Neoplásicas/virologia , Regiões Promotoras Genéticas , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Especificidade da Espécie , Transcrição Gênica
3.
Retrovirology ; 11: 94, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25365920

RESUMO

BACKGROUND: Formation of a mature core is a crucial event for infectivity of retroviruses such as Mason-Pfizer monkey virus (M-PMV). The process is triggered by proteolytic cleavage of the polyprotein precursor Gag, which releases matrix, capsid (CA), and nucleocapsid proteins. Once released, CA assembles to form a mature core - a hexameric lattice protein shell that protects retroviral genomic RNA. Subtle conformational changes within CA induce the transition from the immature lattice to the mature lattice. Upon release from the precursor, the initially unstructured N-terminus of CA is refolded to form a ß-hairpin stabilized by a salt bridge between the N-terminal proline and conserved aspartate. Although the crucial role of the ß-hairpin in the mature core assembly has been confirmed, its precise structural function remains poorly understood. RESULTS: Based on a previous NMR analysis of the N-terminal part of M-PMV CA, which suggested the role of additional interactions besides the proline-aspartate salt bridge in stabilization of the ß-hairpin, we introduced a series of mutations into the CA sequence. The effect of the mutations on virus assembly and infectivity was analyzed. In addition, the structural consequences of selected mutations were determined by NMR spectroscopy. We identified a network of interactions critical for proper formation of the M-PMV core. This network involves residue R14, located in the N-terminal ß-hairpin; residue W52 in the loop connecting helices 2 and 3; and residues Q113, Q115, and Y116 in helix 5. CONCLUSION: Combining functional and structural analyses, we identified a network of supportive interactions that stabilize the ß-hairpin in mature M-PMV CA.


Assuntos
Proteínas do Capsídeo/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Estrutura Secundária de Proteína/genética , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Células HEK293 , Humanos , Vírus dos Macacos de Mason-Pfizer/genética , Dados de Sequência Molecular , Mutação/genética , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Vírion/genética , Vírion/metabolismo , Montagem de Vírus/genética
4.
Virology ; 449: 109-19, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24418544

RESUMO

The intracellular transport of Mason-Pfizer monkey virus (M-PMV) assembled capsids from the pericentriolar region to the plasma membrane (PM) requires trafficking of envelope glycoprotein (Env) to the assembly site via the recycling endosome. However, it is unclear if Env-containing vesicles play a direct role in trafficking capsids to the PM. Using live cell microscopy, we demonstrate, for the first time, anterograde co-transport of Gag and Env. Nocodazole disruption of microtubules had differential effects on Gag and Env trafficking, with pulse-chase assays showing a delayed release of Env-deficient virions. Particle tracking demonstrated an initial loss of linear movement of GFP-tagged capsids and mCherry-tagged Env, followed by renewed movement of Gag but not Env at 4h post-treatment. Thus, while delayed capsid trafficking can occur in the absence of microtubules, efficient anterograde transport of capsids appears to be mediated by microtubule-associated Env-containing vesicles.


Assuntos
Produtos do Gene env/metabolismo , Produtos do Gene gag/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Microtúbulos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Animais , Membrana Celular/virologia , Chlorocebus aethiops , Produtos do Gene env/genética , Produtos do Gene gag/genética , Macaca mulatta , Vírus dos Macacos de Mason-Pfizer/genética , Microtúbulos/metabolismo , Transporte Proteico , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo
5.
PLoS One ; 8(12): e83863, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386297

RESUMO

Immature capsids of the Betaretrovirus, Mason-Pfizer Monkey virus (M-PMV), are assembled in the pericentriolar region of the cell, and are then transported to the plasma membrane for budding. Although several studies, utilizing mutagenesis, biochemistry, and immunofluorescence, have defined the role of some viral and host cells factors involved in these processes, they have the disadvantage of population analysis, rather than analyzing individual capsid movement in real time. In this study, we created an M-PMV vector in which the enhanced green fluorescent protein, eGFP, was fused to the carboxyl-terminus of the M-PMV Gag polyprotein, to create a Gag-GFP fusion that could be visualized in live cells. In order to express this fusion protein in the context of an M-PMV proviral backbone, it was necessary to codon-optimize gag, optimize the Kozak sequence preceding the initiating methionine, and mutate an internal methionine codon to one for alanine (M100A) to prevent internal initiation of translation. Co-expression of this pSARM-Gag-GFP-M100A vector with a WT M-PMV provirus resulted in efficient assembly and release of capsids. Results from fixed-cell immunofluorescence and pulse-chase analyses of wild type and mutant Gag-GFP constructs demonstrated comparable intracellular localization and release of capsids to untagged counterparts. Real-time, live-cell visualization and analysis of the GFP-tagged capsids provided strong evidence for a role for microtubules in the intracellular transport of M-PMV capsids. Thus, this M-PMV Gag-GFP vector is a useful tool for identifying novel virus-cell interactions involved in intracellular M-PMV capsid transport in a dynamic, real-time system.


Assuntos
Capsídeo/metabolismo , Produtos do Gene gag/genética , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Vírus dos Macacos de Mason-Pfizer/genética , Microtúbulos/metabolismo , Proteínas Recombinantes de Fusão/genética , Transporte Biológico , Membrana Celular/metabolismo , Sobrevivência Celular , Corantes Fluorescentes/metabolismo , Produtos do Gene gag/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Cinética , Vírus dos Macacos de Mason-Pfizer/metabolismo , Vírus dos Macacos de Mason-Pfizer/fisiologia , Microtúbulos/virologia , Imagem Molecular , Movimento , Transporte Proteico , Provírus/genética , Provírus/metabolismo , Provírus/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Montagem de Vírus
6.
Proc Natl Acad Sci U S A ; 105(30): 10565-70, 2008 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-18647839

RESUMO

Despite extensive data demonstrating that immature retroviral particle assembly can take place either at the plasma membrane or at a distinct location within the cytoplasm, targeting of viral precursor proteins to either assembly site still remains poorly understood. Biochemical data presented here suggest that Tctex-1, a light chain of the molecular motor dynein, is involved in the intracellular targeting of Mason-Pfizer monkey virus (M-PMV) polyproteins to the cytoplasmic assembly site. Comparison of the three-dimensional structures of M-PMV wild-type matrix protein (wt MA) with a single amino acid mutant (R55F), which redirects assembly from a cytoplasmic site to the plasma membrane, revealed different mutual orientations of their C- and N-terminal domains. This conformational change buries a putative intracellular targeting motif located between both domains in the hydrophobic pocket of the MA molecule, thereby preventing the interaction with cellular transport mechanisms.


Assuntos
Membrana Celular/metabolismo , Membrana Celular/virologia , Dineínas/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Nucleares/fisiologia , Retroviridae/metabolismo , Animais , Sítios de Ligação , Transporte Biológico , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , Humanos , Vírus dos Macacos de Mason-Pfizer/metabolismo , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fenótipo , Estrutura Terciária de Proteína , Região do Complexo-t do Genoma
7.
Biomol NMR Assign ; 2(1): 43-5, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19636921

RESUMO

Mason-Pfizer monkey virus (M-PMV) belongs to the family of betaretroviruses characterized by the assembly of immature particles within cytoplasm of infected cells in contrast to other retroviruses (e.g. HIV, RSV) that assemble their immature particles at a plasma membrane. Simultaneously with or shortly after budding a virus-encoded protease is activated and the Gag polyprotein is cleaved into three major structural proteins: matrix (MA), capsid (CA), and nucleocapsid (NC) protein. Mature retroviral CA proteins consist of two independently folded structural domains: N-terminal domain (NTD) and C-terminal dimerization domain (CTD), separated by a flexible linker. As a first step toward the solution structure elucidation, we present nearly complete backbone and side-chain 1H, 13C and 15N resonance assignment of the M-PMV NTD CA.


Assuntos
Proteínas do Capsídeo/química , Espectroscopia de Ressonância Magnética/métodos , Vírus dos Macacos de Mason-Pfizer/metabolismo , Sequência de Aminoácidos , Isótopos de Carbono/química , Dados de Sequência Molecular , Peso Molecular , Isótopos de Nitrogênio/química , Estrutura Terciária de Proteína , Prótons
8.
J Virol ; 80(24): 12102-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17035335

RESUMO

The APOBEC3 protein family can constitute a potent barrier to the successful infection of mammalian species by retroviruses. Therefore, any retrovirus that has evolved the ability to replicate in a given animal must have developed mechanisms that allow it to avoid or inhibit the APOBEC3 proteins expressed in that animal. Here, we demonstrate that Mason-Pfizer monkey virus (MPMV) is resistant to inhibition by the APOBEC3G protein expressed in its normal host, the rhesus macaque, but highly susceptible to inhibition by murine APOBEC3 (mA3). MPMV virion particles fail to package rhesus APOBEC3G (rA3G), and MPMV Gag binds rA3G poorly in coexpressing cells. In contrast, MPMV virions package mA3 efficiently and MPMV Gag-mA3 complexes are readily detected. Moreover, mA3, but not rA3G, partially colocalizes with MPMV Gag in the cytoplasm of coexpressing cells. Previously, we have demonstrated that murine leukemia virus also escapes inhibition by APOBEC3 proteins by avoiding virion incorporation of its cognate APOBEC3 protein, mA3, yet is inhibited by primate APOBEC3G proteins, which it packages effectively (B. P. Doehle, A. Schäfer, H. L. Wiegand, H. P. Bogerd, and B. R. Cullen, J. Virol. 79:8201-8207, 2005). The finding that two essentially unrelated beta- and gammaretroviruses use similar mechanisms to escape inhibition by the APOBEC3 proteins found in their normal host species suggests that the selective exclusion of APOBEC3 proteins from virion particles may be a general mechanism used by simple mammalian retroviruses.


Assuntos
Citidina Desaminase/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Vírion/metabolismo , Montagem de Vírus/genética , Desaminase APOBEC-3G , Animais , Western Blotting , Linhagem Celular Tumoral , Citidina Desaminase/farmacologia , Primers do DNA , Produtos do Gene gag/metabolismo , Humanos , Imunoprecipitação , Macaca mulatta , Vírus dos Macacos de Mason-Pfizer/efeitos dos fármacos , Vírus dos Macacos de Mason-Pfizer/fisiologia , Camundongos , Plasmídeos/genética , Vírion/efeitos dos fármacos , Vírion/genética
9.
Retrovirology ; 3: 62, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16978406

RESUMO

BACKGROUND: Retrovirus particles emerge from the assembly of two structural protein components, Gag that is translated as a soluble protein in the cytoplasm of the host cells, and Env, a type I transmembrane protein. Because both components are translated in different intracellular compartments, elucidating the mechanisms of retrovirus assembly thus requires the study of their intracellular trafficking. RESULTS: We used a CD25 (Tac) chimera-based approach to study the trafficking of Moloney murine leukemia virus and Mason-Pfizer monkey virus Env proteins. We found that the cytoplasmic tails (CTs) of both Env conserved two major signals that control a complex intracellular trafficking. A dileucine-based motif controls the sorting of the chimeras from the trans-Golgi network (TGN) toward endosomal compartments. Env proteins then follow a retrograde transport to the TGN due to the action of a tyrosine-based motif. Mutation of either motif induces the mis-localization of the chimeric proteins and both motifs are found to mediate interactions of the viral CTs with clathrin adaptors. CONCLUSION: This data reveals the unexpected complexity of the intracellular trafficking of retrovirus Env proteins that cycle between the TGN and endosomes. Given that Gag proteins hijack endosomal host proteins, our work suggests that the endosomal pathway may be used by retroviruses to ensure proper encountering of viral structural Gag and Env proteins in cells, an essential step of virus assembly.


Assuntos
Produtos do Gene env/fisiologia , Leucina/química , Vírus da Leucemia Murina/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Tirosina/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Sequência Conservada , Produtos do Gene env/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Receptores de Interleucina-2/biossíntese , Homologia de Sequência de Aminoácidos , Rede trans-Golgi/metabolismo
10.
Virology ; 345(2): 317-27, 2006 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-16297423

RESUMO

The Mason-Pfizer monkey virus (M-PMV) Gag protein follows a morphogenesis pathway in which immature capsids are preassembled within the cytoplasm before interaction with and budding through the plasma membrane. Intracytoplasmic assembly is facilitated by sequences within the p12 domain of Gag that we have termed the Internal Scaffold Domain (ISD). If M-PMV utilizes an ISD then what provides the equivalent function for most other retroviruses that assemble at the plasma membrane? To investigate the possibility that the membrane itself fulfills this role, we have combined functional deletion of the ISD with a mutation that disrupts intracellular targeting or with a plasma membrane targeting signal. By either modification, targeting of ISD-deleted Gag to the plasma membrane restores particle production. These results provide support for a model in which the plasma membrane and the D-type ISD provide an interchangeable scaffold-like function in retrovirus assembly.


Assuntos
Membrana Celular/metabolismo , Produtos do Gene gag/química , Vírus dos Macacos de Mason-Pfizer/metabolismo , Deleção de Sequência , Vírion/metabolismo , Animais , Capsídeo , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Células HeLa , Humanos , Vírus dos Macacos de Mason-Pfizer/genética , Montagem de Vírus
11.
Virology ; 332(2): 659-66, 2005 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-15680431

RESUMO

The Betaretrovirus genus is characterized by the ability to preassemble immature capsids within the cytoplasm. For Mason-Pfizer monkey virus (M-PMV) this ability depends in part upon the unique Internal Scaffold Domain (ISD) within the p12 region of Gag. In this study, we have further characterized the ability of M-PMV p12 to promote Gag-Gag interaction and have examined the Gag polyprotein of the related mouse mammary tumor virus (MMTV) to potentially identify a region with equivalent function. Using the yeast two-hybrid system, we confirmed that both Gag polyproteins strongly interact, primarily through the CA-NC regions, but also through additional domains N-terminal to CA. For M-PMV, this auxiliary interaction domain was p12. For MMTV, no single strongly self-interacting protein was identified. Instead, MMTV Gag appears to utilize the weak contributions of several protein domains to support the main interaction of its CA-NC. Our findings suggest that, in addition to the canonical NC "I-domain" interaction, MMTV Gag self-association results from the concerted action of multiple regions of the polyprotein while M-PMV Gag relies mainly on its p12 domain.


Assuntos
Produtos do Gene gag/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Capsídeo , Primers do DNA , Escherichia coli/genética , Produtos do Gene gag/química , Produtos do Gene gag/genética , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/metabolismo , Dados de Sequência Molecular , Plasmídeos , Reação em Cadeia da Polimerase
12.
Virology ; 317(1): 146-54, 2003 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-14675633

RESUMO

Mason-Pfizer monkey virus (MPMV) and spleen necrosis virus (SNV) are simple retroviruses that encode functionally divergent cis-acting RNA elements that use cellular proteins to facilitate nuclear export and translation of unspliced viral RNA. We tested the hypothesis that a combination of MPMV constitutive transport element (CTE) and SNV or MPMV RU5 translational enhancer on unspliced HIV-1 gag-pol reporter RNA synergistically augments Gag production. Results of transient transfection assays validate the hypothesis of synergistic augmentation in COS cells, but not 293 cells. RNA targeting experiments verified comparable responsiveness to CTE-interactive proteins tethered by RRE and RevM10Tap in COS and 293 cells. Exogeneous expression of Tap and NXT1 was necessary and sufficient to rescue Gag augmentation in 293 cells. Overexpression experiments established that CTE, but not RU5, confers the responsiveness to Tap and NXT1 and that CTE in conjunction with Tap and NXT1 conferred a 30-fold increase in translational utilization of the cytoplasmic RNA. Our results uncovered a previously unidentified role of CTE in conjunction with Tap and NXT1 in commitment to efficient cytoplasmic RNA utilization.


Assuntos
Regiões 5' não Traduzidas/genética , Elementos Facilitadores Genéticos/genética , Gammaretrovirus/genética , Vírus dos Macacos de Mason-Pfizer/genética , Biossíntese de Proteínas , Animais , Células COS , Linhagem Celular , Citoplasma/metabolismo , Proteínas de Fusão gag-pol/genética , Proteínas de Fusão gag-pol/metabolismo , Gammaretrovirus/metabolismo , Regulação da Expressão Gênica , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Genes Reporter/genética , HIV-1/genética , Humanos , Vírus dos Macacos de Mason-Pfizer/metabolismo , RNA Mensageiro , RNA Viral
13.
Virology ; 314(1): 62-73, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-14517060

RESUMO

Retroviral Gag proteins function during early and late stages of the viral life cycle. To gain additional insight into the cellular requirements for viral replication, a two-hybrid screen was used to identify cellular proteins that interact with the Mason-Pfizer monkey virus Gag protein. One of the cellular proteins found was identified as hUbc9, a nuclear pore-associated E2 SUMO conjugating enzyme. In vitro protein interaction assays verified the association and mapped the interaction domain to the CA protein. In vivo, hUbc9 and Gag colocalized in the cytoplasm as discrete foci near the nuclear membrane. In addition, overexpression of hUbc9 in cells caused a fraction of Gag to colocalize with hUbc9 in the nucleus. These experiments demonstrate that hUbc9 and Gag interact in cells, strengthen the hypothesis that Gag proteins transiently associate with the nuclear compartment during viral replication, and suggest that hUbc9 plays a role in this process.


Assuntos
Produtos do Gene gag/metabolismo , Ligases/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Enzimas de Conjugação de Ubiquitina , Animais , Células COS , Citoplasma/metabolismo , Produtos do Gene gag/química , Produtos do Gene gag/genética , Células HeLa , Humanos , Ligases/genética , Precursores de Proteínas/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Replicação Viral
14.
Traffic ; 4(10): 660-70, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12956869

RESUMO

Intracytoplasmic protein targeting in mammalian cells is critical for organelle function as well as virus assembly, but the signals that mediate it are poorly defined. We show here that Mason-Pfizer monkey virus specifically targets Gag precursor proteins to the pericentriolar region of the cytoplasm in a microtubule dependent process through interactions between a short peptide signal, known as the cytoplasmic targeting-retention signal, and the dynein/dynactin motor complex. The Gag molecules are concentrated in pericentriolar microdomains, where they assemble to form immature capsids. Depletion of Gag from this region by cycloheximide treatment, coupled with the presence of ribosomal clusters that are in close vicinity to the assembling capsids, suggests that the dominant N-terminal cytoplasmic targeting-retention signal functions in a cotranslational manner. Transport of the capsids out of the pericentriolar assembly site requires the env-gene product, and a functional vesicular transport system. A single point mutation that renders the cytoplasmic targeting-retention signal defective abrogates pericentriolar targeting of Gag molecules. Thus the previously defined cytoplasmic targeting-retention signal appears to act as a cotranslational intracellular targeting signal that concentrates Gag proteins at the centriole for assembly of capsids.


Assuntos
Centrossomo/metabolismo , Citoplasma/metabolismo , Produtos do Gene gag/metabolismo , Vírus dos Macacos de Mason-Pfizer/fisiologia , Sinais Direcionadores de Proteínas/fisiologia , Animais , Anticorpos Monoclonais/metabolismo , Células COS , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Linhagem Celular , Centrossomo/ultraestrutura , Chlorocebus aethiops , Cicloeximida/farmacologia , Citoplasma/ultraestrutura , Citoplasma/virologia , Produtos do Gene env/metabolismo , Produtos do Gene gag/efeitos dos fármacos , Produtos do Gene gag/genética , Vírus dos Macacos de Mason-Pfizer/metabolismo , Vírus dos Macacos de Mason-Pfizer/ultraestrutura , Microscopia de Fluorescência , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Nocodazol/farmacologia , Mutação Puntual , Processamento de Proteína Pós-Traducional , Sinais Direcionadores de Proteínas/genética , Inibidores da Síntese de Proteínas/farmacologia , Transporte Proteico , Montagem de Vírus
15.
Traffic ; 4(10): 671-80, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12956870

RESUMO

Cytoplasmic transport of Gag molecules to the site of budding is an important but poorly understand process in retroviral assembly. Our previous studies of Mason-Pfizer monkey virus showed that, for this retrovirus, Gag is assembled into capsids at a pericentriolar region and that Env is necessary for efficient transport out of the site. An Env requirement for cytoplasmic transport implicates vesicular trafficking in this process even though the capsids remain cytoplasmic and do not bud into intracellular compartments in the cells studied to date. We show here that the secretory pathway of the cell is not directly involved in Gag transport since the latter was not inhibited by BFA, nor did Gag colocalize with markers of the ER, Golgi, or TGN. Instead, colocalization was observed between Gag and endocytosed transferrin and with Rab11, suggesting that pericentriolar recycling endosomes play a critical role in this process. Mutants of Rab11 that inhibit efflux of transferrin from the recycling endosome also inhibited Gag transport. Our studies show that Env colocalizes with Gag at the pericentriolar assembly site, and provide evidence that Env must travel through this compartment in order to initiate export of the capsids from the site of assembly. Thus, for the first time, endocytic trafficking of a retroviral Env glycoprotein is linked to the efficient cytoplasmic transport of Gag.


Assuntos
Centrossomo/metabolismo , Endossomos/metabolismo , Produtos do Gene env/metabolismo , Produtos do Gene gag/metabolismo , Vírus dos Macacos de Mason-Pfizer/fisiologia , Animais , Brefeldina A/farmacologia , Células COS , Capsídeo/metabolismo , Linhagem Celular , Chlorocebus aethiops , Deleção de Genes , Genes Virais , Cinética , Vírus dos Macacos de Mason-Pfizer/efeitos dos fármacos , Vírus dos Macacos de Mason-Pfizer/metabolismo , Microscopia de Fluorescência , Inibidores da Síntese de Proteínas/farmacologia , Transporte Proteico , Transferrina/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
16.
J Biol Chem ; 278(40): 38803-12, 2003 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-12869552

RESUMO

Betaretroviruses encode dUTPase, an essential factor in DNA metabolism and repair, in the pro open reading frame located between gag and pol. Ribosomal frame-shifts during expression of retroviral proteins provide a unique possibility for covalent joining of nucleocapsid (NC) and dUTPase within Gag-Pro polyproteins. By developing an antibody against the prototype betaretrovirus Mason-Pfizer monkey virus dUTPase, we demonstrate that i) the NC-dUTPase fusion protein exists both within the virions and infected cells providing the only form of dUTPase, and ii) the retroviral protease does not cleave NC-dUTPase either in the virion or in vitro. We show that recombinant betaretroviral NC-dUTPase and dUTPase are both inefficient catalysts compared with all other dUTPases. Dynamic light scattering and gel filtration confirm that the homotrimeric organization, common among dUTPases, is retained in the NC-dUTPase fusion protein. The betaretroviral dUTPase has been crystallized and single crystals contain homotrimers. Oligonucleotide and Zn2+ binding is well retained in the fusion protein, which is the first example of acquisition of a functional nucleic acid binding module by the DNA repair factor dUTPase. Binding of the hexanucleotide ACTGCC or the octanucleotide (TG)4 to NC-dUTPase modulates enzymatic function, indicating that the low catalytic activity may be compensated by adequate localization.


Assuntos
Vírus dos Macacos de Mason-Pfizer/metabolismo , Nucleocapsídeo/metabolismo , Pirofosfatases/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Células COS , Catálise , Linhagem Celular , Cromatografia em Gel , Cromatografia em Camada Fina , Dicroísmo Circular , Clonagem Molecular , Reparo do DNA , Dimerização , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Vetores Genéticos , Cinética , Ligantes , Luz , Modelos Moleculares , Dados de Sequência Molecular , Fases de Leitura Aberta , Plasmídeos/metabolismo , Ligação Proteica , Pirofosfatases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ribossomos/metabolismo , Espalhamento de Radiação , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Espectrofotometria , Difração de Raios X , Raios X
17.
Virology ; 310(2): 310-8, 2003 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-12781718

RESUMO

Processing of Gag polyproteins by viral protease (PR) leads to reorganization of immature retroviral particles and formation of a ribonucleoprotein core. In some retroviruses, such as HIV and RSV, cleavage of a spacer peptide separating capsid and nucleocapsid proteins is essential for the core formation. We show here that no similar spacer peptide is present in the capsid-nucleocapsid (CA-NC) region of Mason-Pfizer monkey virus (M-PMV) and that the CA protein is cleaved in vitro by the PR within the major homology region (MHR) and the NC protein in several sites at the N-terminus. The CA cleavage product was also identified shortly after penetration of M-PMV into COS cells, suggesting that the protease-catalyzed cleavage is involved in core disintegration.


Assuntos
Proteínas do Capsídeo/metabolismo , Endopeptidases/metabolismo , Vírus dos Macacos de Mason-Pfizer/fisiologia , Replicação Viral , Sequência de Aminoácidos , Animais , Células COS , Proteínas do Capsídeo/química , Produtos do Gene gag/metabolismo , Protease de HIV/metabolismo , HIV-1/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Nucleocapsídeo/química , Nucleocapsídeo/metabolismo
18.
J Virol ; 77(14): 7779-85, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12829817

RESUMO

The transmembrane protein of Mason-Pfizer monkey virus contains two heptad repeats that are predicted to form amphipathic alpha-helices that mediate the conformational change necessary for membrane fusion. To analyze the relative sensitivity of the predicted hydrophobic face of the N-terminal heptad repeat to the insertion of uncharged, polar, and charged substitutions, mutations that introduced alanine, serine, or glutamic acid into positions 436, 443, 450, and 457 of the envelope protein were examined. Novel systems using Tat protein and the GHOST cell line were developed to test and quantitate the effects of the mutations on Env-mediated fusion and infectivity of the virus. While no single amino acid change at any of the positions interfered significantly with the synthesis, processing, or transport to the plasma membrane of glycoprotein complexes, 9 of the 12 nonconservative mutations in these residues completely abolished fusion activity and virus infectivity. Mutations in the central positions (443 and 450) of the heptad repeat region were the most detrimental to Env function, and even single alanine substitutions in these positions dramatically altered the fusogenicity of the protein. These results demonstrate that this N-terminal heptad repeat plays a critical role in Env-mediated membrane fusion and highlight the key function of central hydrophobic residues in this process and the sensitivity of all positions to charge substitutions.


Assuntos
Substituição de Aminoácidos , Vírus dos Macacos de Mason-Pfizer/patogenicidade , Fusão de Membrana , Sequências Repetitivas de Aminoácidos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Animais , Células COS , Fusão Celular , Vírus dos Macacos de Mason-Pfizer/genética , Vírus dos Macacos de Mason-Pfizer/metabolismo , Mutagênese Sítio-Dirigida , Sequências Repetidas Terminais , Proteínas do Envelope Viral/genética , Vírion/metabolismo
19.
J Virol ; 77(9): 5192-200, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12692221

RESUMO

Mason-Pfizer monkey virus (M-PMV) encodes a transmembrane (TM) glycoprotein with a 38-amino-acid-long cytoplasmic domain. After the release of the immature virus, a viral protease-mediated cleavage occurs within the cytoplasmic domain, resulting in the loss of 17 amino acids from the carboxy terminus. This maturational cleavage occurs between a histidine at position 21 and a tyrosine at position 22 in the cytoplasmic domain of the TM protein. We have demonstrated previously that a truncated TM glycoprotein with a 21-amino-acid-long cytoplasmic tail showed enhanced fusogenicity but could not be incorporated into virions. These results suggest that postassembly cleavage of the cytoplasmic domain removes a necessary incorporation signal and activates fusion activity. To investigate the contribution of tyrosine residues to the function of the glycoprotein complex and virus replication, we have introduced amino acid substitutions into two tyrosine residues found in the cytoplasmic domain. The effects of these mutations on glycoprotein biosynthesis and function, as well as on virus infectivity, have been examined. Mutation of tyrosine 34 to alanine had little effect on glycoprotein function. In contrast, substitutions at tyrosine 22 modulated fusion activity in either a positive or negative manner, depending on the substituting amino acid. Moreover, any nonaromatic substitution at this position blocked glycoprotein incorporation into virions and abolished infectivity. These results demonstrate that M-PMV employs a tyrosine signal for the selective incorporation of glycoprotein into budding virions. Antibody uptake studies show that tyrosine 22 is part of an efficient internalization signal in the cytoplasmic domain of the M-PMV glycoprotein that can also be positively and negatively influenced by changes at this site.


Assuntos
Regulação Viral da Expressão Gênica , Glicoproteínas/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Tirosina/química , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Fusão Celular , Citoplasma/química , Endocitose , Genes env , Vírus dos Macacos de Mason-Pfizer/química , Vírus dos Macacos de Mason-Pfizer/genética , Vírus dos Macacos de Mason-Pfizer/patogenicidade , Fusão de Membrana , Dados de Sequência Molecular , Mutação , Proteínas do Envelope Viral/genética
20.
J Virol ; 76(21): 10811-20, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12368324

RESUMO

The Mason-Pfizer monkey virus (M-PMV) Gag protein possesses the ability to assemble into an immature capsid when synthesized in a reticulocyte lysate translation system. In contrast, the human immunodeficiency virus (HIV) Gag protein is incapable of assembly in parallel assays. To enable the assembly of HIV Gag, we have combined or inserted regions of M-PMV Gag into HIV Gag. By both biochemical and morphological criteria, several of these chimeric Gag molecules are capable of assembly into immature capsid-like structures in this in vitro system. Chimeric species containing large regions of M-PMV Gag fused to HIV Gag sequences failed to assemble, while species consisting of only the M-PMV p12 region, and its internal scaffold domain (ISD), fused to HIV Gag were capable of assembly, albeit at reduced kinetics compared to M-PMV Gag. The ability of the ISD to induce assembly of HIV Gag, which normally assembles at the plasma membrane, suggests a common requirement for a concentrating factor in retrovirus assembly. Despite the dramatic effect of the ISD on chimera assembly, the function of HIV Gag domains in that process was found to remain essential, since an assembly-defective mutant of HIV CA, M185A, abolished assembly when introduced into the chimera. This continued requirement for HIV Gag domain function in the assembly of chimeric molecules will allow this in vitro system to be used for the analysis of potential inhibitors of HIV immature particle assembly.


Assuntos
Produtos do Gene gag/metabolismo , HIV-1/metabolismo , Vírus dos Macacos de Mason-Pfizer/metabolismo , Montagem de Vírus , Animais , Capsídeo , Produtos do Gene gag/genética , Genes Virais , HIV-1/fisiologia , Humanos , Cinética , Vírus dos Macacos de Mason-Pfizer/genética , Mutagênese Insercional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA