Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 22(10): e3002827, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39361708

RESUMO

The packaging of genomic RNA (gRNA) into retroviral particles relies on the specific recognition by the Gag precursor of packaging signals (Psi), which maintain a complex secondary structure through long-range interactions (LRIs). However, it remains unclear whether the binding of Gag to Psi alone is enough to promote RNA packaging and what role LRIs play in this process. Using mouse mammary tumor virus (MMTV), we investigated the effects of mutations in 4 proposed LRIs on gRNA structure and function. Our findings revealed the presence of an unsuspected extended LRI, and hSHAPE revealed that maintaining a wild-type-like Psi structure is crucial for efficient packaging. Surprisingly, filter-binding assays demonstrated that most mutants, regardless of their packaging capability, exhibited significant binding to Pr77Gag, suggesting that Gag binding to Psi is insufficient for efficient packaging. Footprinting experiments indicated that efficient RNA packaging is promoted when Pr77Gag binds to 2 specific sites within Psi, whereas binding elsewhere in Psi does not lead to efficient packaging. Taken together, our results suggest that the 3D structure of the Psi/Pr77Gag complex regulates the assembly of viral particles around gRNA, enabling effective discrimination against other viral and cellular RNAs that may also bind Gag efficiently.


Assuntos
Produtos do Gene gag , Vírus do Tumor Mamário do Camundongo , RNA Viral , Montagem de Vírus , RNA Viral/metabolismo , RNA Viral/genética , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/metabolismo , Animais , Produtos do Gene gag/metabolismo , Produtos do Gene gag/genética , Camundongos , Conformação de Ácido Nucleico , Humanos , Ligação Proteica , Mutação , Empacotamento do Genoma Viral , Células HEK293
2.
RNA ; 30(1): 68-88, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-37914398

RESUMO

The retroviral Gag precursor plays a central role in the selection and packaging of viral genomic RNA (gRNA) by binding to virus-specific packaging signal(s) (psi or ψ). Previously, we mapped the feline immunodeficiency virus (FIV) ψ to two discontinuous regions within the 5' end of the gRNA that assumes a higher order structure harboring several structural motifs. To better define the region and structural elements important for gRNA packaging, we methodically investigated these FIV ψ sequences using genetic, biochemical, and structure-function relationship approaches. Our mutational analysis revealed that the unpaired U85CUG88 stretch within FIV ψ is crucial for gRNA encapsidation into nascent virions. High-throughput selective 2' hydroxyl acylation analyzed by primer extension (hSHAPE) performed on wild type (WT) and mutant FIV ψ sequences, with substitutions in the U85CUG88 stretch, revealed that these mutations had limited structural impact and maintained nucleotides 80-92 unpaired, as in the WT structure. Since these mutations dramatically affected packaging, our data suggest that the single-stranded U85CUG88 sequence is important during FIV RNA packaging. Filter-binding assays performed using purified FIV Pr50Gag on WT and mutant U85CUG88 ψ RNAs led to reduced levels of Pr50Gag binding to mutant U85CUG88 ψ RNAs, indicating that the U85CUG88 stretch is crucial for ψ RNA-Pr50Gag interactions. Delineating sequences important for FIV gRNA encapsidation should enhance our understanding of both gRNA packaging and virion assembly, making them potential targets for novel retroviral therapeutic interventions, as well as the development of FIV-based vectors for human gene therapy.


Assuntos
Vírus da Imunodeficiência Felina , Animais , Gatos , Humanos , Vírus da Imunodeficiência Felina/genética , Vírus da Imunodeficiência Felina/metabolismo , RNA Guia de Sistemas CRISPR-Cas , RNA Viral/química , Sítios de Ligação , Genômica , Montagem de Vírus/genética
3.
Nucleic Acids Res ; 51(12): 6479-6494, 2023 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-37224537

RESUMO

A fundamental step in the influenza A virus (IAV) replication cycle is the coordinated packaging of eight distinct genomic RNA segments (i.e. vRNAs) into a viral particle. Although this process is thought to be controlled by specific vRNA-vRNA interactions between the genome segments, few functional interactions have been validated. Recently, a large number of potentially functional vRNA-vRNA interactions have been detected in purified virions using the RNA interactome capture method SPLASH. However, their functional significance in coordinated genome packaging remains largely unclear. Here, we show by systematic mutational analysis that mutant A/SC35M (H7N7) viruses lacking several prominent SPLASH-identified vRNA-vRNA interactions involving the HA segment package the eight genome segments as efficiently as the wild-type virus. We therefore propose that the vRNA-vRNA interactions identified by SPLASH in IAV particles are not necessarily critical for the genome packaging process, leaving the underlying molecular mechanism elusive.


Assuntos
Vírus da Influenza A Subtipo H7N7 , Empacotamento do Genoma Viral , Humanos , Genoma Viral , Vírus da Influenza A Subtipo H7N7/fisiologia , Influenza Humana/virologia , RNA Viral/metabolismo , Montagem de Vírus
4.
Nucleic Acids Res ; 50(16): 9023-9038, 2022 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-35993811

RESUMO

The genome of influenza A virus (IAV) consists of eight unique viral RNA segments. This genome organization allows genetic reassortment between co-infecting IAV strains, whereby new IAVs with altered genome segment compositions emerge. While it is known that reassortment events can create pandemic IAVs, it remains impossible to anticipate reassortment outcomes with pandemic prospects. Recent research indicates that reassortment is promoted by a viral genome packaging mechanism that delivers the eight genome segments as a supramolecular complex into the virus particle. This finding holds promise of predicting pandemic IAVs by understanding the intermolecular interactions governing this genome packaging mechanism. Here, we critically review the prevailing mechanistic model postulating that IAV genome packaging is orchestrated by a network of intersegmental RNA-RNA interactions. Although we find supporting evidence, including segment-specific packaging signals and experimentally proposed RNA-RNA interaction networks, this mechanistic model remains debatable due to a current shortage of functionally validated intersegmental RNA-RNA interactions. We speculate that identifying such functional intersegmental RNA-RNA contacts might be hampered by limitations of the utilized probing techniques and the inherent complexity of the genome packaging mechanism. Nevertheless, we anticipate that improved probing strategies combined with a mutagenesis-based validation could facilitate their discovery.


Assuntos
Vírus da Influenza A , Influenza Humana , Humanos , Empacotamento do Genoma Viral , Montagem de Vírus/genética , Vírus da Influenza A/genética , RNA Viral/genética , Genoma Viral/genética
5.
Nucleic Acids Res ; 49(8): 4668-4688, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33836091

RESUMO

Retroviral RNA genome (gRNA) harbors cis-acting sequences that facilitate its specific packaging from a pool of other viral and cellular RNAs by binding with high-affinity to the viral Gag protein during virus assembly. However, the molecular intricacies involved during selective gRNA packaging are poorly understood. Binding and footprinting assays on mouse mammary tumor virus (MMTV) gRNA with purified Pr77Gag along with in cell gRNA packaging study identified two Pr77Gag binding sites constituting critical, non-redundant packaging signals. These included: a purine loop in a bifurcated stem-loop containing the gRNA dimerization initiation site, and the primer binding site (PBS). Despite these sites being present on both unspliced and spliced RNAs, Pr77Gag specifically bound to unspliced RNA, since only that could adopt the native bifurcated stem-loop structure containing looped purines. These results map minimum structural elements required to initiate MMTV gRNA packaging, distinguishing features that are conserved amongst divergent retroviruses from those perhaps unique to MMTV. Unlike purine-rich motifs frequently associated with packaging signals, direct involvement of PBS in gRNA packaging has not been documented in retroviruses. These results enhance our understanding of retroviral gRNA packaging/assembly, making it not only a target for novel therapeutic interventions, but also development of safer gene therapy vectors.


Assuntos
Produtos do Gene gag/metabolismo , Vírus do Tumor Mamário do Camundongo/metabolismo , Splicing de RNA , RNA Viral/metabolismo , Montagem de Vírus/genética , Animais , Sítios de Ligação/genética , Primers do DNA , Difusão Dinâmica da Luz , Produtos do Gene gag/genética , Genoma Viral , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Conformação de Ácido Nucleico , Purinas , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real
6.
RNA Biol ; 19(1): 191-205, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35067194

RESUMO

Maturation of the HIV-1 viral particles shortly after budding is required for infectivity. During this process, the Pr55Gag precursor undergoes a cascade of proteolytic cleavages, and whilst the structural rearrangements of the viral proteins are well understood, the concomitant maturation of the genomic RNA (gRNA) structure is unexplored, despite evidence that it is required for infectivity. To get insight into this process, we systematically analysed the interactions between Pr55Gag or its maturation products (NCp15, NCp9 and NCp7) and the 5' gRNA region and their structural consequences, in vitro. We show that Pr55Gag and its maturation products mostly bind at different RNA sites and with different contributions of their two zinc knuckle domains. Importantly, these proteins have different transient and permanent effects on the RNA structure, the late NCp9 and NCp7 inducing dramatic structural rearrangements. Altogether, our results reveal the distinct contributions of the different Pr55Gag maturation products on the gRNA structural maturation.


Assuntos
Regiões 5' não Traduzidas , Regulação Viral da Expressão Gênica , Infecções por HIV/virologia , HIV-1/fisiologia , RNA Viral/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Conformação de Ácido Nucleico , Ligação Proteica , Processamento de Proteína Pós-Traducional , RNA Viral/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Montagem de Vírus , Replicação Viral
7.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33799890

RESUMO

Protein post-translational modifications (PTMs) play key roles in eukaryotes since they finely regulate numerous mechanisms used to diversify the protein functions and to modulate their signaling networks. Besides, these chemical modifications also take part in the viral hijacking of the host, and also contribute to the cellular response to viral infections. All domains of the human immunodeficiency virus type 1 (HIV-1) Gag precursor of 55-kDa (Pr55Gag), which is the central actor for viral RNA specific recruitment and genome packaging, are post-translationally modified. In this review, we summarize the current knowledge about HIV-1 Pr55Gag PTMs such as myristoylation, phosphorylation, ubiquitination, sumoylation, methylation, and ISGylation in order to figure out how these modifications affect the precursor functions and viral replication. Indeed, in HIV-1, PTMs regulate the precursor trafficking between cell compartments and its anchoring at the plasma membrane, where viral assembly occurs. Interestingly, PTMs also allow Pr55Gag to hijack the cell machinery to achieve viral budding as they drive recognition between viral proteins or cellular components such as the ESCRT machinery. Finally, we will describe and compare PTMs of several other retroviral Gag proteins to give a global overview of their role in the retroviral life cycle.


Assuntos
HIV-1/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , RNA Viral/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Membrana Celular/metabolismo , Membrana Celular/virologia , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , HIV-1/genética , Humanos , Precursores de Proteínas/genética , RNA Viral/genética , Montagem de Vírus/genética , Replicação Viral/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
8.
Biophys J ; 119(2): 419-433, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32574557

RESUMO

The human immunodeficiency virus type 1 Gag precursor specifically selects the unspliced viral genomic RNA (gRNA) from the bulk of cellular and spliced viral RNAs via its nucleocapsid (NC) domain and drives gRNA encapsidation at the plasma membrane (PM). To further identify the determinants governing the intracellular trafficking of Gag-gRNA complexes and their accumulation at the PM, we compared, in living and fixed cells, the interactions between gRNA and wild-type Gag or Gag mutants carrying deletions in NC zinc fingers (ZFs) or a nonmyristoylated version of Gag. Our data showed that the deletion of both ZFs simultaneously or the complete NC domain completely abolished intracytoplasmic Gag-gRNA interactions. Deletion of either ZF delayed the delivery of gRNA to the PM but did not prevent Gag-gRNA interactions in the cytoplasm, indicating that the two ZFs display redundant roles in this respect. However, ZF2 played a more prominent role than ZF1 in the accumulation of the ribonucleoprotein complexes at the PM. Finally, the myristate group, which is mandatory for anchoring the complexes at the PM, was found to be dispensable for the association of Gag with the gRNA in the cytosol.


Assuntos
HIV-1 , Membrana Celular , Genômica , HIV-1/genética , Humanos , RNA Guia de Cinetoplastídeos , RNA Viral , Montagem de Vírus , Dedos de Zinco
9.
Nucleic Acids Res ; 46(9): e57, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29514260

RESUMO

Non-coding RNA regulatory elements are important for viral replication, making them promising targets for therapeutic intervention. However, regulatory RNA is challenging to detect and characterise using classical structure-function assays. Here, we present in cell Mutational Interference Mapping Experiment (in cell MIME) as a way to define RNA regulatory landscapes at single nucleotide resolution under native conditions. In cell MIME is based on (i) random mutation of an RNA target, (ii) expression of mutated RNA in cells, (iii) physical separation of RNA into functional and non-functional populations, and (iv) high-throughput sequencing to identify mutations affecting function. We used in cell MIME to define RNA elements within the 5' region of the HIV-1 genomic RNA (gRNA) that are important for viral replication in cells. We identified three distinct RNA motifs controlling intracellular gRNA production, and two distinct motifs required for gRNA packaging into virions. Our analysis reveals the 73AAUAAA78 polyadenylation motif within the 5' PolyA domain as a dual regulator of gRNA production and gRNA packaging, and demonstrates that a functional polyadenylation signal is required for viral packaging even though it negatively affects gRNA production.


Assuntos
HIV-1/genética , RNA Viral/biossíntese , RNA Viral/química , Sequências Reguladoras de Ácido Ribonucleico , Montagem de Vírus , Regiões 5' não Traduzidas , Genoma Viral , Células HEK293 , HIV-1/fisiologia , Humanos , Mutação , Motivos de Nucleotídeos , Poli A/metabolismo , Replicação Viral
10.
Virologie (Montrouge) ; 24(6): 381-418, 2020 12 01.
Artigo em Francês | MEDLINE | ID: mdl-33441290

RESUMO

The innate immune response is nonspecific and constitutes the first line of defense against infections by pathogens, mainly by enabling their elimination by phagocytosis or apoptosis. In immune cells, this response is characterized, amongst others, by the synthesis of restriction factors, a class of proteins whose role is to inhibit viral replication. Among them, the proteins of the APOBEC3 (Apolipoprotein B mRNA-editing Enzyme Catalytic polypeptide-like 3 or A3) family are major antiviral factors that target a wide range of viruses. One of their targets is the Human Immunodeficiency Virus Type 1 (HIV-1): the deaminase activity of some A3 proteins converts a fraction of cytidines of the viral genome into uridines, impairing its expression. Nevertheless, HIV-1 counteracts A3 proteins thanks to its Vif protein, which inhibits them by hijacking several cellular mechanisms. Besides, APOBEC3 proteins help maintaining the genome integrity by inhibiting retroelements but they also contribute to carcinogenesis, as it is the case for A3A and A3B, two major factors in this process. The large range of A3 activities, combined with recent studies showing their implication in the regulation of emerging viruses (Zika, SARS-CoV-2), allow A3 and their viral partners to be considered as therapeutic areas.


Assuntos
Desaminases APOBEC/fisiologia , COVID-19/imunologia , Imunidade Inata , Adulto , Motivos de Aminoácidos , Animais , Proteínas de Ciclo Celular/metabolismo , Citidina Desaminase/fisiologia , Reparo do DNA , DNA Viral/metabolismo , Desaminação , Humanos , Mamíferos/metabolismo , MicroRNAs/genética , Modelos Moleculares , Terapia de Alvo Molecular , Mutagênese , Neoplasias/enzimologia , Neoplasias/etiologia , Neoplasias/genética , Prognóstico , Conformação Proteica , Edição de RNA , Relação Estrutura-Atividade , Transcrição Gênica , Proteínas Virais/metabolismo , Viroses/tratamento farmacológico , Viroses/enzimologia , Viroses/imunologia , Replicação Viral
11.
RNA Biol ; 16(5): 612-625, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30773097

RESUMO

The Mason-Pfizer monkey virus (MPMV) genomic RNA (gRNA) packaging signal is a highly-structured element with several stem-loops held together by two phylogenetically conserved long-range interactions (LRIs) between U5 and gag complementary sequences. These LRIs play a critical role in maintaining the structure of the 5´ end of the MPMV gRNA. Thus, one could hypothesize that the overall RNA secondary structure of this region is further architecturally held together by three other stem loops (SL3, Gag SL1, and Gag SL2) comprising of sequences from the distal parts of the 5´untranslated region (5' UTR) to ~ 120 nucleotides into gag, excluding gag sequences involved in forming the U5-Gag LRIs. To provide functional evidence for the biological significance of these stem loops during gRNA encapsidation, these structural motifs were mutated and their effects on MPMV RNA packaging and propagation were tested in a single round trans-complementation assay. The mutant RNA structures were further studied by high throughput SHAPE (hSHAPE) assay. Our results reveal that sequences involved in forming these three stem loops do not play crucial roles at an individual level during MPMV gRNA packaging or propagation. Further structure-function analysis indicates that the U5-Gag LRIs have a more important architectural role in stabilizing the higher order structure of the 5´ UTR than the three stem loops which have a more secondary and perhaps indirect role in stabilizing the overall RNA secondary structure of the region. Our work provides a better understanding of the molecular interactions that take place during MPMV gRNA packaging.


Assuntos
Produtos do Gene gag/genética , Vírus dos Macacos de Mason-Pfizer/fisiologia , RNA Viral/química , RNA Viral/genética , Regiões 5' não Traduzidas , Produtos do Gene gag/química , Humanos , Vírus dos Macacos de Mason-Pfizer/genética , Modelos Moleculares , Mutação , Conformação de Ácido Nucleico , Estabilidade de RNA , Montagem de Vírus
12.
Nat Methods ; 12(9): 866-72, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26237229

RESUMO

RNA regulates many biological processes; however, identifying functional RNA sequences and structures is complex and time-consuming. We introduce a method, mutational interference mapping experiment (MIME), to identify, at single-nucleotide resolution, the primary sequence and secondary structures of an RNA molecule that are crucial for its function. MIME is based on random mutagenesis of the RNA target followed by functional selection and next-generation sequencing. Our analytical approach allows the recovery of quantitative binding parameters and permits the identification of base-pairing partners directly from the sequencing data. We used this method to map the binding site of the human immunodeficiency virus-1 (HIV-1) Pr55(Gag) protein on the viral genomic RNA in vitro, and showed that, by analyzing permitted base-pairing patterns, we could model RNA structure motifs that are crucial for protein binding.


Assuntos
Mutagênese Sítio-Dirigida/métodos , Precursores de Proteínas/química , Precursores de Proteínas/genética , RNA Viral/química , RNA Viral/genética , Análise de Sequência de RNA/métodos , Sequência de Bases , Dados de Sequência Molecular , Mutação/genética , Relação Estrutura-Atividade
13.
RNA ; 22(6): 905-19, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27095024

RESUMO

MPMV has great potential for development as a vector for gene therapy. In this respect, precisely defining the sequences and structural motifs that are important for dimerization and packaging of its genomic RNA (gRNA) are of utmost importance. A distinguishing feature of the MPMV gRNA packaging signal is two phylogenetically conserved long-range interactions (LRIs) between U5 and gag complementary sequences, LRI-I and LRI-II. To test their biological significance in the MPMV life cycle, we introduced mutations into these structural motifs and tested their effects on MPMV gRNA packaging and propagation. Furthermore, we probed the structure of key mutants using SHAPE (selective 2'hydroxyl acylation analyzed by primer extension). Disrupting base-pairing of the LRIs affected gRNA packaging and propagation, demonstrating their significance to the MPMV life cycle. A double mutant restoring a heterologous LRI-I was fully functional, whereas a similar LRI-II mutant failed to restore gRNA packaging and propagation. These results demonstrate that while LRI-I acts at the structural level, maintaining base-pairing is not sufficient for LRI-II function. In addition, in vitro RNA dimerization assays indicated that the loss of RNA packaging in LRI mutants could not be attributed to the defects in dimerization. Our findings suggest that U5-gag LRIs play an important architectural role in maintaining the structure of the 5' region of the MPMV gRNA, expanding the crucial role of LRIs to the nonlentiviral group of retroviruses.


Assuntos
Genes gag , Vírus dos Macacos de Mason-Pfizer/genética , RNA Viral/genética , Montagem de Vírus
14.
RNA Biol ; 15(8): 1047-1059, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29929424

RESUMO

Packaging the mouse mammary tumor virus (MMTV) genomic RNA (gRNA) requires the entire 5' untranslated region (UTR) in conjunction with the first 120 nucleotides of the gag gene. This region includes several palindromic (pal) sequence(s) and stable stem loops (SLs). Among these, stem loop 4 (SL4) adopts a bifurcated structure consisting of three stems, two apical loops, and an internal loop. Pal II, located in one of the apical loops, mediates gRNA dimerization, a process intricately linked to packaging. We thus hypothesized that the bifurcated SL4 structure could constitute the major gRNA packaging determinant. To test this hypothesis, the two apical loops and the flanking sequences forming the bifurcated SL4 were individually mutated. These mutations all had deleterious effects on gRNA packaging and propagation. Next, single and compensatory mutants were designed to destabilize then recreate the bifurcated SL4 structure. A structure-function analysis using bioinformatics predictions and RNA chemical probing revealed that mutations that led to the loss of the SL4 bifurcated structure abrogated RNA packaging and propagation, while compensatory mutations that recreated the native SL4 structure restored RNA packaging and propagation to wild type levels. Altogether, our results demonstrate that SL4 constitutes the principal packaging determinant of MMTV gRNA. Our findings further suggest that SL4 acts as a structural switch that can not only differentiate between RNA for translation versus packaging/dimerization, but its location also allows differentiation between spliced and unspliced RNAs during gRNA encapsidation.


Assuntos
Dimerização , Vírus do Tumor Mamário do Camundongo/metabolismo , Biossíntese de Proteínas , RNA Viral/química , RNA Viral/metabolismo , Montagem de Vírus , Animais , Genômica , Vírus do Tumor Mamário do Camundongo/química , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Conformação de Ácido Nucleico , RNA Viral/genética
15.
RNA Biol ; 15(7): 923-936, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29954247

RESUMO

The Pr55Gag precursor specifically selects the HIV-1 genomic RNA (gRNA) from a large excess of cellular and partially or fully spliced viral RNAs and drives the virus assembly at the plasma membrane. During these processes, the NC domain of Pr55Gag interacts with the gRNA, while its C-terminal p6 domain binds cellular and viral factors and orchestrates viral particle release. Gag∆p6 is a truncated form of Pr55Gag lacking the p6 domain usually used as a default surrogate for wild type Pr55Gag for in vitro analysis. With recent advance in production of full-length recombinant Pr55Gag, here, we tested whether the p6 domain also contributes to the RNA binding specificity of Pr55Gag by systematically comparing binding of Pr55Gag and Gag∆p6 to a panel of viral and cellular RNAs. Unexpectedly, our fluorescence data reveal that the p6 domain is absolutely required for specific binding of Pr55Gag to the HIV-1 gRNA. Its deletion resulted not only in a decreased affinity for gRNA, but also in an increased affinity for spliced viral and cellular RNAs. In contrast Gag∆p6 displayed a similar affinity for all tested RNAs. Removal of the C-terminal His-tag from Pr55Gag and Gag∆p6 uniformly increased the Kd values of the RNA-protein complexes by ~ 2.5 fold but did not affect the binding specificities of these proteins. Altogether, our results demonstrate a novel role of the p6 domain in the specificity of Pr55Gag-RNA interactions, and strongly suggest that the p6 domain contributes to the discrimination of HIV-1 gRNA from cellular and spliced viral mRNAs, which is necessary for its selective encapsidation.


Assuntos
Genoma Viral/genética , HIV-1/fisiologia , Precursores de Proteínas/metabolismo , RNA Viral/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Sítios de Ligação , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , HIV-1/genética , Humanos , Mutação , Plasmídeos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Precursores de Proteínas/genética , RNA Viral/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
16.
Bioinformatics ; 32(21): 3369-3370, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27402903

RESUMO

The mutational interference mapping experiment (MIME) is a powerful method that, coupled to a bioinformatics analysis pipeline, allows the identification of domains and structures in RNA that are important for its function. In MIME, target RNAs are randomly mutated, selected by function, physically separated and sequenced using next-generation sequencing (NGS). Quantitative effects of each mutation at each position in the RNA can be recovered with statistical certainty using the herein developed user-friendly, cross-platform software MIMEAnTo (MIME Analysis Tool). AVAILABILITY AND IMPLEMENTATION: MIMEAnTo is implemented in C ++ using the boost library as well as Qt for the graphical user interface and is distributed under GPL (http://www.gnu.org/licences/gpl). The libraries are statically linked in a stand alone executable and are not required on the system. The plots are generated with gnuplot. Gnuplot-iostream (https://github.com/dstahlke/gnuplot-iostream) serves as gnuplot interface. Standalone executables including examples and source code can be downloaded from https://github.com/maureensmith/MIMEAnTo CONTACTS: msmith@zedat.fu-berlin.de or vkleist@zedat.fu-berlin.deSupplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Mutação , RNA/genética , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Linguagens de Programação , Software
17.
RNA Biol ; 14(1): 90-103, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-27841704

RESUMO

The HIV-1 Pr55Gag precursor specifically selects genomic RNA (gRNA) from a large variety of cellular and spliced viral RNAs (svRNAs), however the molecular mechanisms of this selective recognition remains poorly understood. To gain better understanding of this process, we analyzed the interactions between Pr55Gag and a large panel of viral RNA (vRNA) fragments encompassing the main packaging signal (Psi) and its flanking regions by fluorescence spectroscopy. We showed that the gRNA harbors a high affinity binding site which is absent from svRNA species, suggesting that this site might be crucial for selecting the HIV-1 genome. Our stoichiometry analysis of protein/RNA complexes revealed that few copies of Pr55Gag specifically associate with the 5' region of the gRNA. Besides, we found that gRNA dimerization significantly impacts Pr55Gag binding, and we confirmed that the internal loop of stem-loop 1 (SL1) in Psi is crucial for specific interaction with Pr55Gag. Our analysis of gRNA fragments of different length supports the existence of a long-range tertiary interaction involving sequences upstream and downstream of the Psi region. This long-range interaction might promote optimal exposure of SL1 for efficient Pr55Gag recognition. Altogether, our results shed light on the molecular mechanisms allowing the specific selection of gRNA by Pr55Gag among a variety of svRNAs, all harboring SL1 in their first common exon.


Assuntos
Genoma Viral , HIV-1/genética , HIV-1/metabolismo , Precursores de Proteínas/metabolismo , Splicing de RNA , RNA Viral/genética , RNA Viral/metabolismo , Sítios de Ligação , Humanos , Sequências Repetidas Invertidas , Cinética , Mutação , Conformação de Ácido Nucleico , RNA Viral/química , Proteínas de Ligação a RNA/metabolismo
18.
J Biol Chem ; 290(1): 371-83, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25398876

RESUMO

Because of their high mutation rates, RNA viruses and retroviruses replicate close to the threshold of viability. Their existence as quasi-species has pioneered the concept of "lethal mutagenesis" that prompted us to synthesize pyrimidine nucleoside analogues with antiviral activity in cell culture consistent with an accumulation of deleterious mutations in the HIV-1 genome. However, testing all potentially mutagenic compounds in cell-based assays is tedious and costly. Here, we describe two simple in vitro biophysical/biochemical assays that allow prediction of the mutagenic potential of deoxyribonucleoside analogues. The first assay compares the thermal stabilities of matched and mismatched base pairs in DNA duplexes containing or not the nucleoside analogues as follows. A promising candidate should display a small destabilization of the matched base pair compared with the natural nucleoside and the smallest gap possible between the stabilities of the matched and mismatched base pairs. From this assay, we predicted that two of our compounds, 5-hydroxymethyl-2'-deoxyuridine and 5-hydroxymethyl-2'-deoxycytidine, should be mutagenic. The second in vitro reverse transcription assay assesses DNA synthesis opposite nucleoside analogues inserted into a template strand and subsequent extension of the newly synthesized base pairs. Once again, only 5-hydroxymethyl-2'-deoxyuridine and 5-hydroxymethyl-2'-deoxycytidine are predicted to be efficient mutagens. The predictive potential of our fast and easy first line screens was confirmed by detailed analysis of the mutation spectrum induced by the compounds in cell culture because only compounds 5-hydroxymethyl-2'-deoxyuridine and 5-hydroxymethyl-2'-deoxycytidine were found to increase the mutation frequency by 3.1- and 3.4-fold, respectively.


Assuntos
Fármacos Anti-HIV/química , Transcriptase Reversa do HIV/genética , HIV-1/genética , Ensaios de Triagem em Larga Escala/economia , Mutagênicos/química , Inibidores da Transcriptase Reversa/química , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Pareamento Incorreto de Bases , Pareamento de Bases , Sequência de Bases , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Desenho de Fármacos , Transcriptase Reversa do HIV/antagonistas & inibidores , Transcriptase Reversa do HIV/química , HIV-1/efeitos dos fármacos , HIV-1/enzimologia , Dados de Sequência Molecular , Mutagênese , Mutagênicos/metabolismo , Mutagênicos/farmacologia , Desnaturação de Ácido Nucleico , Valor Preditivo dos Testes , Inibidores da Transcriptase Reversa/metabolismo , Inibidores da Transcriptase Reversa/farmacologia , Transcrição Reversa , Termodinâmica , Timidina/análogos & derivados , Timidina/química , Timidina/metabolismo , Timidina/farmacologia , Fatores de Tempo
19.
Proc Natl Acad Sci U S A ; 110(41): 16604-9, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24067651

RESUMO

Influenza A viruses cause annual influenza epidemics and occasional severe pandemics. Their genome is segmented into eight fragments, which offers evolutionary advantages but complicates genomic packaging. The existence of a selective packaging mechanism, in which one copy of each viral RNA is specifically packaged into each virion, is suspected, but its molecular details remain unknown. Here, we identified a direct intermolecular interaction between two viral genomic RNA segments of an avian influenza A virus using in vitro experiments. Using silent trans-complementary mutants, we then demonstrated that this interaction takes place in infected cells and is required for optimal viral replication. Disruption of this interaction did not affect the HA titer of the mutant viruses, suggesting that the same amount of viral particles was produced. However, it nonspecifically decreased the amount of viral RNA in the viral particles, resulting in an eightfold increase in empty viral particles. Competition experiments indicated that this interaction favored copackaging of the interacting viral RNA segments. The interaction we identified involves regions not previously designated as packaging signals and is not widely conserved among influenza A virus. Combined with previous studies, our experiments indicate that viral RNA segments can promote the selective packaging of the influenza A virus genome by forming a sequence-dependent supramolecular network of interactions. The lack of conservation of these interactions might limit genetic reassortment between divergent influenza A viruses.


Assuntos
Vírus da Influenza A Subtipo H5N2/genética , RNA Viral/metabolismo , Montagem de Vírus/fisiologia , Replicação Viral/fisiologia , Animais , Sequência de Bases , Clonagem Molecular , Análise por Conglomerados , Primers do DNA/genética , Cães , Eletroforese em Gel de Ágar , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H5N2/fisiologia , Células Madin Darby de Rim Canino , Microscopia Eletrônica de Transmissão , Modelos Genéticos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Filogenia , Plasmídeos/genética , RNA Viral/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Montagem de Vírus/genética
20.
Proc Natl Acad Sci U S A ; 110(40): E3840-8, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043788

RESUMO

The fragmented nature of the influenza A genome allows the exchange of gene segments when two or more influenza viruses infect the same cell, but little is known about the rules underlying this process. Here, we studied genetic reassortment between the A/Moscow/10/99 (H3N2, MO) virus originally isolated from human and the avian A/Finch/England/2051/91 (H5N2, EN) virus and found that this process is strongly biased. Importantly, the avian HA segment never entered the MO genetic background alone but always was accompanied by the avian PA and M fragments. Introduction of the 5' and 3' packaging sequences of HA(MO) into an otherwise HA(EN) backbone allowed efficient incorporation of the chimerical viral RNA (vRNA) into the MO genetic background. Furthermore, forcing the incorporation of the avian M segment or introducing five silent mutations into the human M segment was sufficient to drive coincorporation of the avian HA segment into the MO genetic background. These silent mutations also strongly affected the genotype of reassortant viruses. Taken together, our results indicate that packaging signals are crucial for genetic reassortment and that suboptimal compatibility between the vRNA packaging signals, which are detected only when vRNAs compete for packaging, limit this process.


Assuntos
Aves/virologia , Coinfecção/virologia , Transferência Genética Horizontal/genética , Hemaglutininas Virais/genética , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H5N2/genética , Montagem de Vírus/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA/genética , Cães , Genótipo , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Dados de Sequência Molecular , Mutação/genética , Análise de Sequência de DNA , Transdução de Sinais/genética , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA