Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Retrovirology ; 21(1): 13, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898526

RESUMO

Retroviruses exploit host proteins to assemble and release virions from infected cells. Previously, most studies focused on interacting partners of retroviral Gag proteins that localize to the cytoplasm or plasma membrane. Given that several full-length Gag proteins have been found in the nucleus, identifying the Gag-nuclear interactome has high potential for novel findings involving previously unknown host processes. Here we systematically compared nuclear factors identified in published HIV-1 proteomic studies and performed our own mass spectrometry analysis using affinity-tagged HIV-1 and RSV Gag proteins mixed with nuclear extracts. We identified 57 nuclear proteins in common between HIV-1 and RSV Gag, and a set of nuclear proteins present in our analysis and ≥ 1 of the published HIV-1 datasets. Many proteins were associated with nuclear processes which could have functional consequences for viral replication, including transcription initiation/elongation/termination, RNA processing, splicing, and chromatin remodeling. Examples include facilitating chromatin remodeling to expose the integrated provirus, promoting expression of viral genes, repressing the transcription of antagonistic cellular genes, preventing splicing of viral RNA, altering splicing of cellular RNAs, or influencing viral or host RNA folding or RNA nuclear export. Many proteins in our pulldowns common to RSV and HIV-1 Gag are critical for transcription, including PolR2B, the second largest subunit of RNA polymerase II (RNAPII), and LEO1, a PAF1C complex member that regulates transcriptional elongation, supporting the possibility that Gag influences the host transcription profile to aid the virus. Through the interaction of RSV and HIV-1 Gag with splicing-related proteins CBLL1, HNRNPH3, TRA2B, PTBP1 and U2AF1, we speculate that Gag could enhance unspliced viral RNA production for translation and packaging. To validate one putative hit, we demonstrated an interaction of RSV Gag with Mediator complex member Med26, required for RNA polymerase II-mediated transcription. Although 57 host proteins interacted with both Gag proteins, unique host proteins belonging to each interactome dataset were identified. These results provide a strong premise for future functional studies to investigate roles for these nuclear host factors that may have shared functions in the biology of both retroviruses, as well as functions specific to RSV and HIV-1, given their distinctive hosts and molecular pathology.


Assuntos
Produtos do Gene gag , HIV-1 , Humanos , HIV-1/fisiologia , HIV-1/genética , Produtos do Gene gag/metabolismo , Produtos do Gene gag/genética , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Vírus do Sarcoma de Rous/fisiologia , Vírus do Sarcoma de Rous/genética , Proteômica , Interações Hospedeiro-Patógeno , Replicação Viral , Interações entre Hospedeiro e Microrganismos , Espectrometria de Massas
2.
Viruses ; 13(9)2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34578434

RESUMO

The small cellular molecule inositol hexakisphosphate (IP6) has been known for ~20 years to promote the in vitro assembly of HIV-1 into immature virus-like particles. However, the molecular details underlying this effect have been determined only recently, with the identification of the IP6 binding site in the immature Gag lattice. IP6 also promotes formation of the mature capsid protein (CA) lattice via a second IP6 binding site, and enhances core stability, creating a favorable environment for reverse transcription. IP6 also enhances assembly of other retroviruses, from both the Lentivirus and the Alpharetrovirus genera. These findings suggest that IP6 may have a conserved function throughout the family Retroviridae. Here, we discuss the different steps in the viral life cycle that are influenced by IP6, and describe in detail how IP6 interacts with the immature and mature lattices of different retroviruses.


Assuntos
HIV-1/fisiologia , Ácido Fítico/metabolismo , Retroviridae/fisiologia , Montagem de Vírus , Sítios de Ligação , Proteínas do Capsídeo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Mutação , Proteínas dos Retroviridae/metabolismo , Transcrição Reversa , Vírus do Sarcoma de Rous/fisiologia , Replicação Viral , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
3.
Nat Commun ; 12(1): 3226, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34050170

RESUMO

Inositol hexakisphosphate (IP6) is an assembly cofactor for HIV-1. We report here that IP6 is also used for assembly of Rous sarcoma virus (RSV), a retrovirus from a different genus. IP6 is ~100-fold more potent at promoting RSV mature capsid protein (CA) assembly than observed for HIV-1 and removal of IP6 in cells reduces infectivity by 100-fold. Here, visualized by cryo-electron tomography and subtomogram averaging, mature capsid-like particles show an IP6-like density in the CA hexamer, coordinated by rings of six lysines and six arginines. Phosphate and IP6 have opposing effects on CA in vitro assembly, inducing formation of T = 1 icosahedrons and tubes, respectively, implying that phosphate promotes pentamer and IP6 hexamer formation. Subtomogram averaging and classification optimized for analysis of pleomorphic retrovirus particles reveal that the heterogeneity of mature RSV CA polyhedrons results from an unexpected, intrinsic CA hexamer flexibility. In contrast, the CA pentamer forms rigid units organizing the local architecture. These different features of hexamers and pentamers determine the structural mechanism to form CA polyhedrons of variable shape in mature RSV particles.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/ultraestrutura , Ácido Fítico/metabolismo , Vírus do Sarcoma de Rous/ultraestrutura , Montagem de Vírus , Capsídeo/metabolismo , Proteínas do Capsídeo/isolamento & purificação , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Modelos Moleculares , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Multimerização Proteica , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Vírus do Sarcoma de Rous/patogenicidade , Vírus do Sarcoma de Rous/fisiologia , Imagem Individual de Molécula , Transfecção , Liberação de Vírus
4.
J Virol ; 94(17)2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32581109

RESUMO

Retroviral Gag polyproteins orchestrate the assembly and release of nascent virus particles from the plasma membranes of infected cells. Although it was traditionally thought that Gag proteins trafficked directly from the cytosol to the plasma membrane, we discovered that the oncogenic avian alpharetrovirus Rous sarcoma virus (RSV) Gag protein undergoes transient nucleocytoplasmic transport as an intrinsic step in virus assembly. Using a genetic approach in yeast, we identified three karyopherins that engage the two independent nuclear localization signals (NLSs) in Gag. The primary NLS is in the nucleocapsid (NC) domain of Gag and binds directly to importin-α, which recruits importin-ß to mediate nuclear entry. The second NLS (TNPO3), which resides in the matrix (MA) domain, is dependent on importin-11 and transportin-3 (TNPO3), which are known as MTR10p and Kap120p in yeast, although it is not clear whether these import factors are independent or additive. The functions of importin-α/importin-ß and importin-11 have been verified in avian cells, whereas the role of TNPO3 has not been studied. In this report, we demonstrate that TNPO3 directly binds to Gag and mediates its nuclear entry. To our surprise, this interaction did not require the cargo-binding domain (CBD) of TNPO3, which typically mediates nuclear entry for other binding partners of TNPO3, including SR domain-containing splicing factors and tRNAs that reenter the nucleus. These results suggest that RSV hijacks this host nuclear import pathway using a unique mechanism, potentially allowing other cargo to simultaneously bind TNPO3.IMPORTANCE RSV Gag nuclear entry is facilitated using three distinct host import factors that interact with nuclear localization signals in the Gag MA and NC domains. Here, we show that the MA region is required for nuclear import of Gag through the TNPO3 pathway. Gag nuclear entry does not require the CBD of TNPO3. Understanding the molecular basis for TNPO3-mediated nuclear trafficking of the RSV Gag protein may lead to a deeper appreciation for whether different import factors play distinct roles in retrovirus replication.


Assuntos
Produtos do Gene gag/metabolismo , Domínios Proteicos , Vírus do Sarcoma de Rous/fisiologia , Internalização do Vírus , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , Núcleo Celular , Produtos do Gene gag/genética , Carioferinas/metabolismo , Sinais de Localização Nuclear/metabolismo , Nucleocapsídeo/metabolismo , Transporte Proteico , Saccharomyces cerevisiae , Montagem de Vírus , alfa Carioferinas/metabolismo , beta Carioferinas/genética
5.
Viruses ; 12(4)2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32230826

RESUMO

The Rous sarcoma virus Gag polyprotein transiently traffics through the nucleus, which is required for efficient incorporation of the viral genomic RNA (gRNA) into virus particles. Packaging of gRNA is mediated by two zinc knuckles and basic residues located in the nucleocapsid (NC) domain in Gag. To further examine the role of basic residues located downstream of the zinc knuckles in gRNA encapsidation, we used a gain-of-function approach. We replaced a basic residue cluster essential for gRNA packaging with heterologous basic residue motif (BR) with RNA-binding activity from either the HIV-1 Rev protein (Rev BR) or the HSV ICP27 protein (ICP27 BR). Compared to wild-type Gag, the mutant ICP27 BR and Rev BR Gag proteins were much more strongly localized to the nucleus and released significantly lower levels of virus particles. Surprisingly, both the ICP27 BR and Rev BR mutants packaged normal levels of gRNA per virus particle when examined in the context of a proviral vector, yet both mutants were noninfectious. These results support the hypothesis that basic residues located in the C-terminal region of NC are required for selective gRNA packaging, potentially by binding non-specifically to RNA via electrostatic interactions.


Assuntos
Substituição de Aminoácidos , Produtos do Gene gag/química , Produtos do Gene gag/genética , Motivos de Ligação ao RNA , Vírus do Sarcoma de Rous/fisiologia , Proteínas Virais/química , Proteínas Virais/genética , Produtos do Gene gag/metabolismo , Genoma Viral , Humanos , Ligação Proteica , Transporte Proteico , Proteínas Virais/metabolismo , Montagem de Vírus , Liberação de Vírus
6.
Front Biosci (Landmark Ed) ; 24(3): 463-481, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30468667

RESUMO

The present study determines the cytokine gene expression in chickens following RSV-A infection, using RT-qPCR. In susceptible chickens tumors progressed to  fulminating metastatic tumors while it regressed in  regressors  chickens and some resistant non-responder chickens did not respond to RSV-A infection and thus did not develop tumors at all. The in vivo expression of pro-inflammatory cytokines, Th1 cytokines and Th2 cytokines was determined at the primary site of infection, as well as in different organs of progressor, regressor and non-responder chicks at different time intervals. Our results indicated a significant upregulation of the pro-inflammatory cytokines, IL-6 and IL-8, in all the organs of progressor chicks, while they were significantly lower in regressor and non-responder chicks. The expression of the Th1 cytokines IFN-γ and TNF-α was low in all of the organs of the progressor group, except that in  spleen. In contrast, regressor and non-responder groups showed high expression of IFN-γ and TNF-α. Further, there was an early upregulation of the expression of the Th2 cytokine, IL-10, TGF-ß and GM-CSF, in all of the organs of progressors as compared to uninfected control.


Assuntos
Citocinas/imunologia , Vírus do Sarcoma de Rous/imunologia , Sarcoma Aviário/imunologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Galinhas , Citocinas/genética , Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/imunologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Vírus do Sarcoma de Rous/fisiologia , Sarcoma Aviário/genética , Sarcoma Aviário/virologia , Células Th1/metabolismo , Células Th1/virologia , Células Th2/metabolismo , Células Th2/virologia , Regulação para Cima/imunologia
7.
Subcell Biochem ; 88: 211-243, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29900499

RESUMO

Integration of the reverse-transcribed viral cDNA into the host's genome is a critical step in the lifecycle of all retroviruses. Retrovirus integration is carried out by integrase (IN), a virus-encoded enzyme that forms an oligomeric 'intasome' complex with both ends of the linear viral DNA to catalyze their concerted insertions into the backbones of the host's DNA. IN also forms a complex with host proteins, which guides the intasome to the host's chromosome. Recent structural studies have revealed remarkable diversity as well as conserved features among the architectures of the intasome assembly from different genera of retroviruses. This chapter will review how IN oligomerizes to achieve its function, with particular focus on alpharetrovirus including the avian retrovirus Rous sarcoma virus. Another chapter (Craigie) will focus on the structure and function of IN from HIV-1.


Assuntos
DNA Complementar , DNA Viral , Integrases , Vírus do Sarcoma de Rous , Proteínas Virais , Integração Viral/fisiologia , Animais , DNA Complementar/química , DNA Complementar/genética , DNA Complementar/metabolismo , DNA Viral/química , DNA Viral/genética , DNA Viral/metabolismo , Humanos , Integrases/genética , Integrases/metabolismo , Vírus do Sarcoma de Rous/química , Vírus do Sarcoma de Rous/fisiologia , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
8.
Viruses ; 10(4)2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29670049

RESUMO

The Czech scientist Jan Svoboda was a pioneer of Rous sarcoma virus (RSV). In the 1960s, before the discovery of reverse transcriptase, he demonstrated the long-term persistence of the viral genome in non-productive mammalian cells, and he supported the DNA provirus hypothesis of Howard Temin. He showed how the virus can be rescued in the infectious form and elucidated the replication-competent nature of the Prague strain of RSV later used for the identification of the src oncogene. His studies straddled molecular oncology and virology, and he remained an active contributor to the field until his death last year. Throughout the 50 years that I was privileged to know Svoboda as my mentor and friend, I admired his depth of scientific inquiry and his steadfast integrity in the face of political oppression.


Assuntos
Interações Hospedeiro-Patógeno , Vírus do Sarcoma de Rous/fisiologia , Vírus do Sarcoma de Rous/patogenicidade , Sarcoma Aviário/virologia , Replicação Viral , Animais , História do Século XX , História do Século XXI , Humanos
9.
Biochim Biophys Acta Gene Regul Mech ; 1861(4): 387-400, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29155070

RESUMO

Prokaryotic and eukaryotic cytosolic aminoacyl-tRNA synthetases (aaRSs) are essentially known for their conventional function of generating the full set of aminoacyl-tRNA species that are needed to incorporate each organism's repertoire of genetically-encoded amino acids during ribosomal translation of messenger RNAs. However, bacterial and eukaryotic cytosolic aaRSs have been shown to exhibit other essential nonconventional functions. Here we review all the subcellular compartments that prokaryotic and eukaryotic cytosolic aaRSs can reach to exert either a conventional or nontranslational role. We describe the physiological and stress conditions, the mechanisms and the signaling pathways that trigger their relocation and the new functions associated with these relocating cytosolic aaRS. Finally, given that these relocating pools of cytosolic aaRSs participate to a wide range of cellular pathways beyond translation, but equally important for cellular homeostasis, we mention some of the pathologies and diseases associated with the dis-regulation or malfunctioning of these nontranslational functions.


Assuntos
Aminoácidos/metabolismo , Aminoacil-tRNA Sintetases/fisiologia , Citosol/enzimologia , RNA de Transferência/metabolismo , Aminoacilação de RNA de Transferência/fisiologia , Aminoacil-tRNA Sintetases/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Transporte Biológico , Citocinas/biossíntese , Células Eucarióticas/enzimologia , HIV/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana/fisiologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/fisiologia , Proteínas de Neoplasias/fisiologia , Neovascularização Fisiológica/fisiologia , Fagocitose/fisiologia , Células Procarióticas/enzimologia , Isoformas de Proteínas/fisiologia , Vírus do Sarcoma de Rous/fisiologia , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiologia , Especificidade da Espécie , Vertebrados/genética , Vertebrados/metabolismo
10.
Sci Rep ; 7(1): 2913, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28588198

RESUMO

During a proteolytically-driven maturation process, the orthoretroviral capsid protein (CA) assembles to form the convex shell that surrounds the viral genome. In some orthoretroviruses, including Rous Sarcoma Virus (RSV), CA carries a short and hydrophobic spacer peptide (SP) at its C-terminus early in the maturation process, which is progressively removed as maturation proceeds. In this work, we show that RSV CA assembles in vitro at near-physiological temperatures, forming hexamer tubes that effectively model the mature capsid surface. Tube assembly is strongly influenced by electrostatic effects, and is a nucleated process that remains thermodynamically favored at lower temperatures, but is effectively arrested by the large Gibbs energy barrier associated with nucleation. RSV CA tubes are multi-layered, being formed by nested and concentric tubes of capsid hexamers. However the spacer peptide acts as a layering determinant during tube assembly. If only a minor fraction of CA-SP is present, multi-layered tube formation is blocked, and single-layered tubes predominate. This likely prevents formation of biologically aberrant multi-layered capsids in the virion. The generation of single-layered hexamer tubes facilitated 3D helical image reconstruction from cryo-electron microscopy data, revealing the basic tube architecture.


Assuntos
Proteínas do Capsídeo/metabolismo , Vírus do Sarcoma de Rous/fisiologia , Montagem de Vírus , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/ultraestrutura , Imageamento Tridimensional , Técnicas In Vitro , Modelos Moleculares , Concentração Osmolar , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Proteólise , Eletricidade Estática , Temperatura
11.
J Biol Chem ; 292(12): 5018-5030, 2017 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-28184005

RESUMO

The retrovirus integrase (IN) inserts the viral cDNA into the host DNA genome. Atomic structures of five different retrovirus INs complexed with their respective viral DNA or branched viral/target DNA substrates have indicated these intasomes are composed of IN subunits ranging from tetramers, to octamers, or to hexadecamers. IN precursors are monomers, dimers, or tetramers in solution. But how intasome assembly is controlled remains unclear. Therefore, we sought to unravel the functional mechanisms in different intasomes. We produced kinetically stabilized Rous sarcoma virus (RSV) intasomes with human immunodeficiency virus type 1 strand transfer inhibitors that interact simultaneously with IN and viral DNA within intasomes. We examined the ability of RSV IN dimers to assemble two viral DNA molecules into intasomes containing IN tetramers in contrast to one possessing IN octamers. We observed that the last 18 residues of the C terminus ("tail" region) of IN (residues 1-286) determined whether an IN tetramer or octamer assembled with viral DNA. A series of truncations of the tail region indicated that these 18 residues are critical for the assembly of an intasome containing IN octamers but not for an intasome containing IN tetramers. The C-terminally truncated IN (residues 1-269) produced an intasome that contained tetramers but failed to produce an intasome with octamers. Both intasomes have similar catalytic activities. The results suggest a high degree of plasticity for functional multimerization and reveal a critical role of the C-terminal tail region of IN in higher order oligomerization of intasomes, potentially informing future strategies to prevent retroviral integration.


Assuntos
DNA Viral/metabolismo , Integrases/metabolismo , Vírus do Sarcoma de Rous/enzimologia , Animais , Aves , Cristalografia por Raios X , Humanos , Integrases/química , Modelos Moleculares , Multimerização Proteica , Vírus do Sarcoma de Rous/química , Vírus do Sarcoma de Rous/fisiologia , Sarcoma Aviário/metabolismo , Sarcoma Aviário/virologia , Integração Viral
12.
Trends Biochem Sci ; 41(5): 410-420, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27039020

RESUMO

Retroviral capsid cores are proteinaceous containers that self-assemble to encase the viral genome and a handful of proteins that promote infection. Their function is to protect and aid in the delivery of viral genes to the nucleus of the host, and, in many cases, infection pathways are influenced by capsid-cellular interactions. From a mathematical perspective, capsid cores are polyhedral cages and, as such, follow well-defined geometric rules. However, marked morphological differences in shapes exist, depending on virus type. Given the specific roles of capsid in the viral life cycle, the availability of detailed molecular structures, particularly at assembly interfaces, opens novel avenues for targeted drug development against these pathogens. Here, we summarize recent advances in the structure and understanding of retroviral capsid, with particular emphasis on assemblies and the capsid cores.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/ultraestrutura , HIV-1/ultraestrutura , Vírus da Leucemia Bovina/ultraestrutura , Vírus do Sarcoma de Rous/ultraestrutura , Vírion/ultraestrutura , Sítios de Ligação , Capsídeo/química , Capsídeo/fisiologia , Proteínas do Capsídeo/metabolismo , Cristalografia por Raios X , HIV-1/química , HIV-1/fisiologia , Vírus da Leucemia Bovina/química , Vírus da Leucemia Bovina/fisiologia , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estrutura Terciária de Proteína , Vírus do Sarcoma de Rous/química , Vírus do Sarcoma de Rous/fisiologia , Vírion/química , Vírion/fisiologia , Montagem de Vírus
13.
J Virol ; 90(12): 5700-5714, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27053549

RESUMO

UNLABELLED: Extensive studies of orthoretroviral capsids have shown that many regions of the CA protein play unique roles at different points in the virus life cycle. The N-terminal domain (NTD) flexible-loop (FL) region is one such example: exposed on the outer capsid surface, it has been implicated in Gag-mediated particle assembly, capsid maturation, and early replication events. We have now defined the contributions of charged residues in the FL region of the Rous sarcoma virus (RSV) CA to particle assembly. Effects of mutations on assembly were assessed in vivo and in vitro and analyzed in light of new RSV Gag lattice models. Virus replication was strongly dependent on the preservation of charge at a few critical positions in Gag-Gag interfaces. In particular, a cluster of charges at the beginning of FL contributes to an extensive electrostatic network that is important for robust Gag assembly and subsequent capsid maturation. Second-site suppressor analysis suggests that one of these charged residues, D87, has distal influence on interhexamer interactions involving helix α7. Overall, the tolerance of FL to most mutations is consistent with current models of Gag lattice structures. However, the results support the interpretation that virus evolution has achieved a charge distribution across the capsid surface that (i) permits the packing of NTD domains in the outer layer of the Gag shell, (ii) directs the maturational rearrangements of the NTDs that yield a functional core structure, and (iii) supports capsid function during the early stages of virus infection. IMPORTANCE: The production of infectious retrovirus particles is a complex process, a choreography of protein and nucleic acid that occurs in two distinct stages: formation and release from the cell of an immature particle followed by an extracellular maturation phase during which the virion proteins and nucleic acids undergo major rearrangements that activate the infectious potential of the virion. This study examines the contributions of charged amino acids on the surface of the Rous sarcoma virus capsid protein in the assembly of appropriately formed immature particles and the maturational transitions that create a functional virion. The results provide important biological evidence in support of recent structural models of the RSV immature virions and further suggest that immature particle assembly and virion maturation are controlled by an extensive network of electrostatic interactions and long-range communication across the capsid surface.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Produtos do Gene gag/química , Vírus do Sarcoma de Rous/química , Vírus do Sarcoma de Rous/fisiologia , Montagem de Vírus , Sequência de Aminoácidos , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Produtos do Gene gag/genética , Microscopia Eletrônica , Modelos Moleculares , Mutação , Vírus do Sarcoma de Rous/genética , Vírus do Sarcoma de Rous/ultraestrutura , Eletricidade Estática , Vírion/metabolismo , Vírion/ultraestrutura
14.
J Virol ; 90(5): 2473-85, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26676779

RESUMO

UNLABELLED: The principles underlying membrane binding and assembly of retroviral Gag proteins into a lattice are understood. However, little is known about how these processes are related. Using purified Rous sarcoma virus Gag and Gag truncations, we studied the interrelation of Gag-Gag interaction and Gag-membrane interaction. Both by liposome binding and by surface plasmon resonance on a supported bilayer, Gag bound to membranes much more tightly than did matrix (MA), the isolated membrane binding domain. In principle, this difference could be explained either by protein-protein interactions leading to cooperativity in membrane binding or by the simultaneous interaction of the N-terminal MA and the C-terminal nucleocapsid (NC) of Gag with the bilayer, since both are highly basic. However, we found that NC was not required for strong membrane binding. Instead, the spacer peptide assembly domain (SPA), a putative 24-residue helical sequence comprising the 12-residue SP segment of Gag and overlapping the capsid (CA) C terminus and the NC N terminus, was required. SPA is known to be critical for proper assembly of the immature Gag lattice. A single amino acid mutation in SPA that abrogates assembly in vitro dramatically reduced binding of Gag to liposomes. In vivo, plasma membrane localization was dependent on SPA. Disulfide cross-linking based on ectopic Cys residues showed that the contacts between Gag proteins on the membrane are similar to the known contacts in virus-like particles. Taken together, we interpret these results to mean that Gag membrane interaction is cooperative in that it depends on the ability of Gag to multimerize. IMPORTANCE: The retroviral structural protein Gag has three major domains. The N-terminal MA domain interacts directly with the plasma membrane (PM) of cells. The central CA domain, together with immediately adjoining sequences, facilitates the assembly of thousands of Gag molecules into a lattice. The C-terminal NC domain interacts with the genome, resulting in packaging of viral RNA. For assembly in vitro with purified Gag, in the absence of membranes, binding of NC to nucleic acid somehow facilitates further Gag-Gag interactions that lead to formation of the Gag lattice. The contributions of MA-mediated membrane binding to virus particle assembly are not well understood. Here, we report that in the absence of nucleic acid, membranes provide a platform that facilitates Gag-Gag interactions. This study demonstrates that the binding of Gag, but not of MA, to membranes is cooperative and identifies SPA as a major factor that controls this cooperativity.


Assuntos
Produtos do Gene gag/metabolismo , Bicamadas Lipídicas/metabolismo , Multimerização Proteica , Vírus do Sarcoma de Rous/fisiologia , Análise Mutacional de DNA , Produtos do Gene gag/genética , Ligação Proteica , Estrutura Terciária de Proteína , Vírus do Sarcoma de Rous/genética
15.
Folia Biol (Praha) ; 61(5): 161-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26667572

RESUMO

In my article I tried to present the results of early experiments suggesting a significant role for cell association in Rous sarcoma virus transformation of non-permissive cells and revealing that infectious virus can be efficiently rescued from such cells by their fusion with permissive chicken fibroblasts.


Assuntos
Galinhas/virologia , Vírus do Sarcoma de Rous/patogenicidade , Sarcoma Aviário/virologia , Animais , Transformação Celular Viral , Provírus/patogenicidade , Provírus/fisiologia , Ratos , Vírus do Sarcoma de Rous/fisiologia , Replicação Viral
16.
Virus Res ; 193: 78-88, 2014 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-25016037

RESUMO

The Gag precursor of HIV-1, formed of the four proteic regions matrix (MA), capsid (CA), nucleocapsid (NC) and p6, orchestrates virus morphogenesis. This complex process relies on three major interactions, NC-RNA acting as a scaffold, CA-CA and MA-membrane that targets assembly to the plasma membrane (PM). The characterization of the molecular mechanism of retroviral assembly has extensively benefited from biochemical studies and more recently an important step forward was achieved with the use of fluorescence-based techniques and fluorescently labeled viral proteins. In this review, we summarize the findings obtained with such techniques, notably quantitative-based approaches, which highlight the role of the NC region in Gag assembly.


Assuntos
HIV-1/fisiologia , Nucleocapsídeo/metabolismo , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Animais , Membrana Celular/metabolismo , Humanos , Microscopia de Fluorescência , Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/metabolismo , Ligação Proteica , Multimerização Proteica , Transporte Proteico , Vírus do Sarcoma de Rous/fisiologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química
17.
J Biol Chem ; 289(28): 19648-58, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24872410

RESUMO

We determined conditions to produce milligram quantities of the soluble Rous sarcoma virus (RSV) synaptic complex that is kinetically trapped by HIV strand transfer inhibitors (STIs). Concerted integration catalyzed by RSV integrase (IN) is effectively inhibited by HIV STIs. Optimized assembly of the RSV synaptic complex required IN, a gain-of-function 3'-OH-recessed U3 oligonucleotide, and an STI under specific conditions to maintain solubility of the trapped synaptic complex at 4 °C. A C-terminal truncated IN (1-269 residues) produced a homogeneous population of trapped synaptic complex that eluted at ∼ 151,000 Da upon Superdex 200 size-exclusion chromatography (SEC). Approximately 90% of input IN and DNA are incorporated into the trapped synaptic complex using either the C-terminally truncated IN or wild type IN (1-286 residues). No STI is present in the SEC running buffer suggesting the STI-trapped synaptic complex is kinetically stabilized. The yield of the trapped synaptic complex correlates with the dissociative half-life of the STI observed with HIV IN-DNA complexes. Dolutegravir, MK-2048, and MK-0536 are equally effective, whereas raltegravir is ∼ 70% as effective. Without an STI present in the assembly mixture, no trapped synaptic complex was observed. Fluorescence and mass spectroscopy analyses demonstrated that the STI remains associated with the trapped complex. SEC-multiangle light scattering analyses demonstrated that wild type IN and the C-terminal IN truncation are dimers that acted as precursors to the tetramer. The purified STI-trapped synaptic complex contained a tetramer as shown by cross-linking studies. Structural studies of this three-domain RSV IN in complex with viral DNA may be feasible.


Assuntos
DNA Viral/química , Integrase de HIV/química , HIV-1/química , Vírus do Sarcoma de Rous/química , DNA Viral/imunologia , Integrase de HIV/metabolismo , HIV-1/fisiologia , Humanos , Estrutura Terciária de Proteína , Vírus do Sarcoma de Rous/fisiologia , Montagem de Vírus/fisiologia
18.
J Virol ; 88(14): 7852-61, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24789780

RESUMO

Host cell tRNAs are recruited for use as primers to initiate reverse transcription in retroviruses. Human immunodeficiency virus type 1 (HIV-1) uses tRNA(Lys3) as the replication primer, whereas Rous sarcoma virus (RSV) uses tRNA(Trp). The nucleic acid (NA) chaperone function of the nucleocapsid (NC) domain of HIV-1 Gag is responsible for annealing tRNA(Lys3) to the genomic RNA (gRNA) primer binding site (PBS). Compared to HIV-1, little is known about the chaperone activity of RSV Gag. In this work, using purified RSV Gag containing an N-terminal His tag and a deletion of the majority of the protease domain (H6.Gag.3h), gel shift assays were used to monitor the annealing of tRNA(Trp) to a PBS-containing RSV RNA. Here, we show that similar to HIV-1 Gag lacking the p6 domain (GagΔp6), RSV H6.Gag.3h is a more efficient chaperone on a molar basis than NC; however, in contrast to the HIV-1 system, both RSV H6.Gag.3h and NC have comparable annealing rates at protein saturation. The NC domain of RSV H6.Gag.3h is required for annealing, whereas deletion of the matrix (MA) domain, which stimulates the rate of HIV-1 GagΔp6 annealing, has little effect on RSV H6.Gag.3h chaperone function. Competition assays confirmed that RSV MA binds inositol phosphates (IPs), but in contrast to HIV-1 GagΔp6, IPs do not stimulate RSV H6.Gag.3h chaperone activity unless the MA domain is replaced with HIV-1 MA. We conclude that differences in the MA domains are primarily responsible for mechanistic differences in RSV and HIV-1 Gag NA chaperone function. Importance: Mounting evidence suggests that the Gag polyprotein is responsible for annealing primer tRNAs to the PBS to initiate reverse transcription in retroviruses, but only HIV-1 Gag chaperone activity has been demonstrated in vitro. Understanding RSV Gag's NA chaperone function will allow us to determine whether there is a common mechanism among retroviruses. This report shows for the first time that full-length RSV Gag lacking the protease domain is a highly efficient NA chaperone in vitro, and NC is required for this activity. In contrast to results obtained for HIV-1 Gag, due to the weak nucleic acid binding affinity of the RSV MA domain, inositol phosphates do not regulate RSV Gag-facilitated tRNA annealing despite the fact that they bind to MA. These studies provide insight into the viral regulation of tRNA primer annealing, which is a potential target for antiretroviral therapy.


Assuntos
Produtos do Gene gag/metabolismo , HIV-1/fisiologia , Chaperonas Moleculares/metabolismo , Fosfoproteínas/metabolismo , RNA de Transferência de Triptofano/metabolismo , RNA Viral/metabolismo , Vírus do Sarcoma de Rous/fisiologia , Proteínas da Matriz Viral/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Antígenos HIV/metabolismo , Humanos , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
19.
J Virol ; 88(13): 7170-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24719425

RESUMO

UNLABELLED: During virion maturation, the Rous sarcoma virus (RSV) capsid protein is cleaved from the Gag protein as the proteolytic intermediate CA-SP. Further trimming at two C-terminal sites removes the spacer peptide (SP), producing the mature capsid proteins CA and CA-S. Abundant genetic and structural evidence shows that the SP plays a critical role in stabilizing hexameric Gag interactions that form immature particles. Freeing of CA-SP from Gag breaks immature interfaces and initiates the formation of mature capsids. The transient persistence of CA-SP in maturing virions and the identification of second-site mutations in SP that restore infectivity to maturation-defective mutant viruses led us to hypothesize that SP may play an important role in promoting the assembly of mature capsids. This study presents a biophysical and biochemical characterization of CA-SP and its assembly behavior. Our results confirm cryo-electron microscopy (cryo-EM) structures reported previously by Keller et al. (J. Virol. 87:13655-13664, 2013, doi:10.1128/JVI.01408-13) showing that monomeric CA-SP is fully capable of assembling into capsid-like structures identical to those formed by CA. Furthermore, SP confers aggressive assembly kinetics, which is suggestive of higher-affinity CA-SP interactions than observed with either of the mature capsid proteins. This aggressive assembly is largely independent of the SP amino acid sequence, but the formation of well-ordered particles is sensitive to the presence of the N-terminal ß-hairpin. Additionally, CA-SP can nucleate the assembly of CA and CA-S. These results suggest a model in which CA-SP, once separated from the Gag lattice, can actively promote the interactions that form mature capsids and provide a nucleation point for mature capsid assembly. IMPORTANCE: The spacer peptide is a documented target for antiretroviral therapy. This study examines the biochemical and biophysical properties of CA-SP, an intermediate form of the retrovirus capsid protein. The results demonstrate a previously unrecognized activity of SP in promoting capsid assembly during maturation.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/metabolismo , Fragmentos de Peptídeos/química , Vírus do Sarcoma de Rous/fisiologia , Montagem de Vírus , Sequência de Aminoácidos , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
20.
J Virol ; 88(10): 5617-29, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24599998

RESUMO

UNLABELLED: Purified retroviral Gag proteins can assemble in vitro to form immature virus-like particles (VLPs). By cryoelectron tomography, Rous sarcoma virus VLPs show an organized hexameric lattice consisting chiefly of the capsid (CA) domain, with periodic stalk-like densities below the lattice. We hypothesize that the structure represented by these densities is formed by amino acid residues immediately downstream of the folded CA, namely, the short spacer peptide SP, along with a dozen flanking residues. These 24 residues comprise the SP assembly (SPA) domain, and we propose that neighboring SPA units in a Gag hexamer coalesce to form a six-helix bundle. Using in vitro assembly, alanine scanning mutagenesis, and biophysical analyses, we have further characterized the structure and function of SPA. Most of the amino acid residues in SPA could not be mutated individually without abrogating assembly, with the exception of a few residues near the N and C termini, as well as three hydrophilic residues within SPA. We interpret these results to mean that the amino acids that do not tolerate mutations contribute to higher-order structures in VLPs. Hydrogen-deuterium exchange analyses of unassembled Gag compared that of assembled VLPs showed strong protection at the SPA region, consistent with a higher-order structure. Circular dichroism revealed that a 29mer SPA peptide shifts from a random coil to a helix in a concentration-dependent manner. Analytical ultracentrifugation showed concentration-dependent self-association of the peptide into a hexamer. Taken together, these results provide strong evidence for the formation of a critical six-helix bundle in Gag assembly. IMPORTANCE: The structure of a retrovirus like HIV is created by several thousand molecules of the viral Gag protein, which assemble to form the known hexagonal protein lattice in the virus particle. How the Gag proteins pack together in the lattice is incompletely understood. A short segment of Gag known to be critical for proper assembly has been hypothesized to form a six-helix bundle, which may be the nucleating event that leads to lattice formation. The experiments reported here, using the avian Rous sarcoma virus as a model system, further define the nature of this segment of Gag, show that it is in a higher-order structure in the virus particle, and provide the first direct evidence that it forms a six-helix bundle in retrovirus assembly. Such knowledge may provide underpinnings for the development of antiretroviral drugs that interfere with virus assembly.


Assuntos
Produtos do Gene gag/metabolismo , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Vírus do Sarcoma de Rous/fisiologia , Montagem de Vírus , Substituição de Aminoácidos , Dicroísmo Circular , Análise Mutacional de DNA , Produtos do Gene gag/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformação Proteica , Vírus do Sarcoma de Rous/genética , Ultracentrifugação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...