Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Retrovirology ; 20(1): 16, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37700325

RESUMO

BACKGROUND: The murine leukemia virus (MLV) has been a powerful model of pathogenesis for the discovery of genes involved in cancer. Its splice donor (SD')-associated retroelement (SDARE) is important for infectivity and tumorigenesis, but the mechanism remains poorly characterized. Here, we show for the first time that P50 protein, which is produced from SDARE, acts as an accessory protein that transregulates transcription and induces cell transformation. RESULTS: By infecting cells with MLV particles containing SDARE transcript alone (lacking genomic RNA), we show that SDARE can spread to neighbouring cells as shown by the presence of P50 in infected cells. Furthermore, a role for P50 in cell transformation was demonstrated by CCK8, TUNEL and anchorage-independent growth assays. We identified the integrase domain of P50 as being responsible for transregulation of the MLV promoter using luciferase assay and RTqPCR with P50 deleted mutants. Transcriptomic analysis furthermore revealed that the expression of hundreds of cellular RNAs involved in cancerogenesis were deregulated in the presence of P50, suggesting that P50 induces carcinogenic processes via its transcriptional regulatory function. CONCLUSION: We propose a novel SDARE-mediated mode of propagation of the P50 accessory protein in surrounding cells. Moreover, due to its transforming properties, P50 expression could lead to a cellular and tissue microenvironment that is conducive to cancer development.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Animais , Genômica , Vírus da Leucemia Murina/genética , Regiões Promotoras Genéticas , RNA
2.
Biophys Rep (N Y) ; 2(3): 100068, 2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36425325

RESUMO

Real-time visualization and quantification of viruses released by a cell are crucial to further decipher infection processes. Kinetics studies at the single-cell level will circumvent the limitations of bulk assays with asynchronous virus replication. We have implemented a "viro-fluidic" method, which combines microfluidics and virology at single-cell and single-virus resolutions. As an experimental model, we used standard cell lines producing fluorescent HIV-like particles (VLPs). First, to scale the strategy to the single-cell level, we validated a sensitive flow virometry system to detect VLPs in low concentration samples (≥104 VLPs/mL). Then, this system was coupled to a single-cell trapping device to monitor in real-time the VLPs released, one at a time, from single cells under cell culture conditions. Our results revealed an average production rate of 50 VLPs/h/cell similar to the rate estimated for the same cells grown in population. Thus, the virus-producing capacities of the trapped cells were preserved and its real-time monitoring was accurate. Moreover, single-cell analysis revealed a release of VLPs with stochastic bursts with typical time intervals of few minutes, revealing the existence of limiting step(s) in the virus biogenesis process. Our tools can be applied to other pathogens or to extracellular vesicles to elucidate the dissemination mechanisms of these biological nanoparticles.

3.
Nano Lett ; 22(9): 3651-3658, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35475610

RESUMO

Nanopores combined with optical approaches can be used to detect viral particles. In this work, we demonstrate the ability of hydrodynamical driving and optical sensing to identify and quantify viral particles in a biological sample. We have developed a simple and rapid method which requires only fluorescent labeling of the particles and can therefore be applied to a wide range of virus type. The system operates in real time and at the single particle level while providing a low error on concentration (4%) and a low limit of detection of 105 particles/mL for an acquisition time of 60 s with the ability to increase the acquisition time to achieve a lower limit.


Assuntos
Vesículas Extracelulares , Nanopartículas , Nanoporos , Vírus , Vírion
4.
J Biol Chem ; 298(1): 101500, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34929171

RESUMO

In HIV, the polyprotein precursor Gag orchestrates the formation of the viral capsid. In the current view of this viral assembly, Gag forms low-order oligomers that bind to the viral genomic RNA triggering the formation of high-ordered ribonucleoprotein complexes. However, this assembly model was established using biochemical or imaging methods that do not describe the cellular location hosting Gag-gRNA complex nor distinguish gRNA packaging in single particles. Here, we studied the intracellular localization of these complexes by electron microscopy and monitored the distances between the two partners by morphometric analysis of gold beads specifically labeling Gag and gRNA. We found that formation of these viral clusters occurred shortly after the nuclear export of the gRNA. During their transport to the plasma membrane, the distance between Gag and gRNA decreases together with an increase of gRNA packaging. Point mutations in the zinc finger patterns of the nucleocapsid domain of Gag caused an increase in the distance between Gag and gRNA as well as a sharp decrease of gRNA packaged into virions. Finally, we show that removal of stem loop 1 of the 5'-untranslated region does not interfere with gRNA packaging, whereas combined with the removal of stem loop 3 is sufficient to decrease but not abolish Gag-gRNA cluster formation and gRNA packaging. In conclusion, this morphometric analysis of Gag-gRNA cluster formation sheds new light on HIV-1 assembly that can be used to describe at nanoscale resolution other viral assembly steps involving RNA or protein-protein interactions.


Assuntos
Produtos do Gene gag , HIV-1 , Nucleoproteínas , Regiões 5' não Traduzidas , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Genômica , HIV-1/genética , HIV-1/metabolismo , Microscopia Eletrônica de Transmissão , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , RNA Guia de Cinetoplastídeos , RNA Viral/genética , RNA Viral/metabolismo , Montagem de Vírus/genética
5.
Viruses ; 12(9)2020 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-32899657

RESUMO

HIV-1 is the causative agent of acquired immunodeficiency syndrome (AIDS). It affects millions of people worldwide and the pandemic persists despite the implementation of highly active antiretroviral therapy. A wide spectrum of techniques has been implemented in order to diagnose and monitor AIDS progression over the years. Besides the conventional approaches, microfluidics has provided useful methods for monitoring HIV-1 infection. In this review, we introduce continuous microfluidics as well as the fabrication and handling of microfluidic chips. We provide a review of the different applications of continuous microfluidics in AIDS diagnosis and progression and in the basic study of the HIV-1 life cycle.


Assuntos
Infecções por HIV/diagnóstico , HIV-1/fisiologia , Técnicas Analíticas Microfluídicas , Microfluídica , Síndrome da Imunodeficiência Adquirida/diagnóstico , Síndrome da Imunodeficiência Adquirida/fisiopatologia , Progressão da Doença , Infecções por HIV/virologia , HIV-1/genética , Humanos , Replicação Viral
6.
J Virol ; 94(18)2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32641479

RESUMO

Apolipoprotein B editing enzyme, catalytic polypeptide 3 (APOBEC3) family members are cytidine deaminases that play important roles in intrinsic responses to retrovirus infection. Complex retroviruses like human immunodeficiency virus type 1 (HIV-1) encode the viral infectivity factor (Vif) protein to counteract APOBEC3 proteins. Vif induces degradation of APOBEC3G and other APOBEC3 proteins and thereby prevents their packaging into virions. It is not known if murine leukemia virus (MLV) encodes a Vif-like protein. Here, we show that the MLV P50 protein, produced from an alternatively spliced gag RNA, interacts with the C terminus of mouse APOBEC3 and prevents its packaging without causing its degradation. By infecting APOBEC3 knockout (KO) and wild-type (WT) mice with Friend or Moloney MLV P50-deficient viruses, we found that APOBEC3 restricts the mutant viruses more than WT viruses in vivo Replication of P50-mutant viruses in an APOBEC3-expressing stable cell line was also much slower than that of WT viruses, and overexpressing P50 in this cell line enhanced mutant virus replication. Thus, MLV encodes a protein, P50, that overcomes APOBEC3 restriction by preventing its packaging into virions.IMPORTANCE MLV has existed in mice for at least a million years, in spite of the existence of host restriction factors that block infection. Although MLV is considered a simple retrovirus compared to lentiviruses, it does encode proteins generated from alternatively spliced RNAs. Here, we show that P50, generated from an alternatively spliced RNA encoded in gag, counteracts APOBEC3 by blocking its packaging. MLV also encodes a protein, glycoGag, that increases capsid stability and limits APOBEC3 access to the reverse transcription complex (RTC). Thus, MLV has evolved multiple means of preventing APOBEC3 from blocking infection, explaining its survival as an infectious pathogen in mice.


Assuntos
Citidina Desaminase/genética , Regulação Viral da Expressão Gênica , Produtos do Gene gag/genética , Leucemia Experimental/genética , Vírus da Leucemia Murina de Moloney/genética , Infecções por Retroviridae/genética , Infecções Tumorais por Vírus/genética , Processamento Alternativo , Animais , Capsídeo/metabolismo , Citidina Desaminase/deficiência , Produtos do Gene gag/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/virologia , Camundongos , Camundongos Knockout , Vírus da Leucemia Murina de Moloney/metabolismo , Vírus da Leucemia Murina de Moloney/patogenicidade , Células NIH 3T3 , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Transdução de Sinais , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/virologia , Vírion/genética , Vírion/metabolismo , Vírion/patogenicidade , Replicação Viral
7.
Nanoscale ; 11(13): 6036-6044, 2019 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-30869094

RESUMO

Membrane partition and remodeling play a key role in numerous cell mechanisms, especially in viral replication cycles where viruses subvert the plasma membrane to enter and escape from the host cell. Specifically assembly and release of HIV-1 particles require specific cellular components, which are recruited to the egress site by the viral protein Gag. We previously demonstrated that HIV-1 assembly alters both partitioning and dynamics of the tetraspanins CD9 and CD81, which are key players in many infectious processes, forming enriched areas where the virus buds. In this study we correlated super resolution microscopy mapping of tetraspanins with membrane topography delineated by atomic force microscopy (AFM) in Gag-expressing cells. We revealed that CD9 is specifically trapped within the nascent viral particles, especially at buds tips, suggesting that Gag mediates CD9 and CD81 depletion from the plasma membrane. In addition, we showed that CD9 is organized as small membrane assemblies of few tens of nanometers that can coalesce upon Gag expression.


Assuntos
HIV-1/fisiologia , Tetraspanina 28/química , Tetraspanina 29/química , Membrana Celular/metabolismo , Citometria de Fluxo , Células HeLa , Humanos , Microscopia de Força Atômica , Tetraspanina 28/metabolismo , Tetraspanina 29/metabolismo , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
8.
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
9.
Viruses ; 9(10)2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28961190

RESUMO

Human immunodeficiency virus (HIV) is a causative agent of acquired immune deficiency syndrome (AIDS). Highly active antiretroviral therapy (HAART) can slow down the replication of HIV-1, leading to an improvement in the survival of HIV-1-infected patients. However, drug toxicities and poor drug administration has led to the emergence of a drug-resistant strain. HIV-1 immunotherapy has been continuously developed, but antibody therapy and HIV vaccines take time to improve its efficiency and have limitations. HIV-1-specific chimeric antigen receptor (CAR)-based immunotherapy founded on neutralizing antibodies is now being developed. In HIV-1 therapy, anti-HIV chimeric antigen receptors showed promising data in the suppression of HIV-1 replication; however, autologous transfusion is still a problem. This has led to the development of effective peptides and proteins for an alternative HIV-1 treatment. In this paper, we provide a comprehensive review of potent anti-HIV-1 peptides and proteins that reveal promising therapeutic activities. The inhibitory mechanisms of each therapeutic molecule in the different stages of the HIV-1 life cycle will be discussed herein.


Assuntos
Terapia Genética , Infecções por HIV/terapia , HIV-1/efeitos dos fármacos , Imunoterapia Adotiva , Peptídeos/uso terapêutico , Proteínas/uso terapêutico , Anticorpos de Cadeia Única/uso terapêutico , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/imunologia , Repetição de Anquirina , Anticorpos Neutralizantes/uso terapêutico , Terapia Antirretroviral de Alta Atividade/efeitos adversos , Farmacorresistência Viral , Anticorpos Anti-HIV/imunologia , Anticorpos Anti-HIV/uso terapêutico , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Integração Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
10.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490595

RESUMO

Along with CD4+ T lymphocytes, macrophages are a major cellular source of HIV-1 replication and a potential viral reservoir. Following entry and reverse transcription in macrophages, cloaking of the viral cDNA by the HIV-1 capsid limits its cytosolic detection, enabling efficient replication. However, whether incoming HIV-1 particles are sensed by macrophages prior to reverse transcription remains unclear. Here, we show that HIV-1 triggers a broad expression of interferon (IFN)-stimulated genes (ISG) in monocyte-derived macrophages within a few hours after infection. This response does not require viral reverse transcription or the presence of HIV-1 RNA within particles, but viral fusion is essential. This response is elicited by viruses carrying different envelope proteins and thus different receptors to proceed for viral entry. Expression of ISG in response to viral entry requires TBK1 activity and type I IFNs signaling. Remarkably, the ISG response is transient but affects subsequent viral spread. Together, our results shed light on an early step of HIV-1 sensing by macrophages at the level of entry, which confers an early protection through type I IFN signaling and has potential implications in controlling the infection.IMPORTANCE HIV infection is restricted to T lymphocytes and macrophages. HIV-1-infected macrophages are found in many tissues of infected patients, even under antiretroviral therapy, and are considered a viral reservoir. How HIV-1 is detected and what type of responses are elicited upon sensing remain in great part elusive. The kinetics and localization of the production of cytokines such as interferons in response to HIV is of critical importance to understanding how the infection and the immune response are established. Our study provides evidence that macrophages can detect HIV-1 as soon as it enters the cell. Interestingly, this sensing is independent of the presence of viral nucleic acids within the particles but requires their fusion with the macrophages. This triggers a low interferon response, which activates an antiviral program protecting cells against further viral challenge and thus potentially limiting the spread of the infection.


Assuntos
HIV-1/imunologia , HIV-1/fisiologia , Imunidade Inata , Interferon Tipo I/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Internalização do Vírus , Células Cultivadas , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Tempo
11.
Viruses ; 8(8)2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27556480

RESUMO

In cells, positive strand RNA viruses, such as Retroviridae, must selectively recognize their full-length RNA genome among abundant cellular RNAs to assemble and release particles. How viruses coordinate the intracellular trafficking of both RNA and protein components to the assembly sites of infectious particles at the cell surface remains a long-standing question. The mechanisms ensuring packaging of genomic RNA are essential for viral infectivity. Since RNA packaging impacts on several essential functions of retroviral replication such as RNA dimerization, translation and recombination events, there are many studies that require the determination of RNA packaging efficiency and/or RNA packaging ability. Studies of RNA encapsidation rely upon techniques for the identification and quantification of RNA species packaged by the virus. This review focuses on the different approaches available to monitor RNA packaging: Northern blot analysis, ribonuclease protection assay and quantitative reverse transcriptase-coupled polymerase chain reaction as well as the most recent RNA imaging and sequencing technologies. Advantages, disadvantages and limitations of these approaches will be discussed in order to help the investigator to choose the most appropriate technique. Although the review was written with the prototypic simple murine leukemia virus (MLV) and complex human immunodeficiency virus type 1 (HIV-1) in mind, the techniques were described in order to benefit to a larger community.


Assuntos
Biologia Molecular/métodos , RNA Viral/metabolismo , Retroviridae/fisiologia , Vírion/metabolismo , Virologia/métodos , Montagem de Vírus , Animais , Humanos
12.
Nucleic Acids Res ; 44(16): 7922-34, 2016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27280976

RESUMO

Dimerization is a unique and vital characteristic of retroviral genomes. It is commonly accepted that genomic RNA (gRNA) must be dimeric at the plasma membrane of the infected cells to be packaged during virus assembly. However, where, when and how HIV-1 gRNA find each other and dimerize in the cell are long-standing questions that cannot be answered using conventional approaches. Here, we combine two state-of-the-art, multicolor RNA labeling strategies with two single-molecule microscopy technologies to address these questions. We used 3D-super-resolution structured illumination microscopy to analyze and quantify the spatial gRNA association throughout the cell and monitored the dynamics of RNA-RNA complexes in living-cells by cross-correlation fluctuation analysis. These sensitive and complementary approaches, combined with trans-complementation experiments, reveal for the first time the presence of interacting gRNA in the cytosol, a challenging observation due to the low frequency of these events and their dilution among the bulk of other RNAs, and allow the determination of the subcellular orchestration of the HIV-1 dimerization process.


Assuntos
Dimerização , HIV-1/genética , Imageamento Tridimensional , Microscopia/métodos , Conformação de Ácido Nucleico , RNA Viral/química , Membrana Celular/metabolismo , Sobrevivência Celular , Cor , Citosol/metabolismo , Genoma Viral , Células HeLa , Humanos , Hibridização in Situ Fluorescente , RNA Viral/genética , Coloração e Rotulagem , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
13.
Sci Rep ; 6: 27536, 2016 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-27273064

RESUMO

HIV-1 is a retrovirus replicating within cells by reverse transcribing its genomic RNA (gRNA) into DNA. Within cells, virus assembly requires the structural Gag proteins with few accessory proteins, notably the viral infectivity factor (Vif) and two copies of gRNA as well as cellular factors to converge to the plasma membrane. In this process, the nucleocapsid (NC) domain of Gag binds to the packaging signal of gRNA which consists of a series of stem-loops (SL1-SL3) ensuring gRNA selection and packaging into virions. Interestingly, mutating NC activates a late-occurring reverse transcription (RT) step in producer cells, leading to the release of DNA-containing HIV-1 particles. In order to decipher the molecular mechanism regulating this late RT, we explored the role of several key partners of NC, such as Vif, gRNA and the cellular cytidine deaminase APOBEC3G that restricts HIV-1 infection by targeting the RT. By studying combinations of deletions of these putative players, we revealed that NC, SL1-SL3 and in lesser extent Vif, but not APOBEC3G, interplay regulates the late RT.


Assuntos
HIV-1/genética , Interações Hospedeiro-Patógeno , Nucleocapsídeo/genética , Transcrição Reversa , Montagem de Vírus/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Desaminase APOBEC-3G/genética , Desaminase APOBEC-3G/metabolismo , Sequência de Bases , Regulação da Expressão Gênica , Células HEK293 , HIV-1/metabolismo , Humanos , Sequências Repetidas Invertidas , Nucleocapsídeo/metabolismo , Ligação Proteica , RNA Guia de Cinetoplastídeos/química , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Deleção de Sequência , Transdução de Sinais , Vírion/genética , Vírion/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo
14.
RNA Biol ; 12(9): 942-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26158194

RESUMO

The retroviral genome consists of an intron-containing transcript that has essential cytoplasmic functions in the infected cell. This viral transcript can escape splicing, circumvent the nuclear checkpoint mechanisms and be transported to the cytoplasm by hijacking the host machinery. Once in the cytoplasm, viral unspliced RNA acts as mRNA to be translated and as genomic RNA to be packaged into nascent viruses. The murine leukemia virus (MLV) is among the first retroviruses discovered and is classified as simple Retroviridae due to its minimal encoding capacity. The oncogenic and transduction abilities of MLV are extensively studied, whereas surprisingly the crucial step of its nuclear export has remained unsolved until 2014. Recent work has revealed the recruitment by MLV of the cellular NXF1/Tap-dependent pathway for export. Unconventionally, MLV uses of Tap to export both spliced and unspliced viral RNAs. Unlike other retroviruses, MLV does not harbor a unique RNA signal for export. Indeed, multiple sequences throughout the MLV genome appear to promote export of the unspliced MLV RNA. We review here the current understanding of the export mechanism and highlight the determinants that influence MLV export. As the molecular mechanism of MLV export is elucidated, we will gain insight into the contribution of the export pathway to the cytoplasmic fate of the viral RNA.


Assuntos
Transporte Ativo do Núcleo Celular , Íntrons , Vírus da Leucemia Murina/fisiologia , RNA Viral/genética , RNA Viral/metabolismo , Processamento Alternativo , Animais , Genoma Viral , Humanos , Sequências Repetidas Invertidas , Camundongos , Conformação de Ácido Nucleico , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Motivos de Nucleotídeos , Fases de Leitura Aberta , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/química , Sequências Reguladoras de Ácido Ribonucleico , Transdução de Sinais , Sequências Repetidas Terminais
15.
Nucleic Acids Res ; 43(1): 336-47, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25488808

RESUMO

HIV-1, the agent of the AIDS pandemic, is an RNA virus that reverse transcribes its RNA genome (gRNA) into DNA, shortly after its entry into cells. Within cells, retroviral assembly requires thousands of structural Gag proteins and two copies of gRNA as well as cellular factors, which converge to the plasma membrane in a finely regulated timeline. In this process, the nucleocapsid domain of Gag (GagNC) ensures gRNA selection and packaging into virions. Subsequent budding and virus release require the recruitment of the cellular ESCRT machinery. Interestingly, mutating GagNC results into the release of DNA-containing viruses, by promo-ting reverse transcription (RTion) prior to virus release, through an unknown mechanism. Therefore, we explored the biogenesis of these DNA-containing particles, combining live-cell total internal-reflection fluorescent microscopy, electron microscopy, trans-complementation assays and biochemical characterization of viral particles. Our results reveal that DNA virus production is the consequence of budding defects associated with Gag aggregation at the plasma membrane and deficiency in the recruitment of Tsg101, a key ESCRT-I component. Indeed, targeting Tsg101 to virus assembly sites restores budding, restricts RTion and favors RNA packaging into viruses. Altogether, our results highlight the role of GagNC in the spatiotemporal control of RTion, via an ESCRT-I-dependent mechanism.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , HIV-1/fisiologia , Fatores de Transcrição/metabolismo , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Membrana Celular/virologia , DNA Viral/biossíntese , HIV-1/genética , HIV-1/metabolismo , Células HeLa , Humanos , Transcrição Reversa , Deleção de Sequência , Vírion/metabolismo , Dedos de Zinco , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química
16.
Retrovirology ; 11: 21, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24597485

RESUMO

BACKGROUND: Eukaryotic cells have evolved stringent proofreading mechanisms to ensure that intron-containing mRNAs do not leave the nucleus. However, all retroviruses must bypass this checkpoint for replication. Indeed, their primary polycistronic transcript (Full-Length) must reach the cytoplasm to be either translated or packaged as genomic RNA in progeny viruses.Murine leukemia virus (MLV) is a prototype of simple retroviruses with only two well-regulated splicing events that directly influence viral leukemogenic properties in mice. Several cis-elements have been identified in the FL RNA that regulate its cytoplasmic accumulation. However, their connection with an export mechanism is yet unknown. Our goal was to identify the cellular pathway used by MLV to export its RNAs into the cytoplasm of the host cells. RESULTS: Since other retroviruses use the CRM1 and/or the Tap/NXF1 pathways to export their unspliced RNA from the nucleus, we investigated the role of these two pathways in MLV replication by using specific inhibitors. The effects of export inhibition on MLV protein synthesis, RNA levels and RNA localization were studied by Western blotting, RT-qPCR, fluorescence microscopy and ribonucleoprotein immunoprecipitation assays. Taken together, our results show for the first time that MLV requires the Tap/NXF1-mediated export pathway, and not the CRM1 pathway, for the expression of its spliced and unspliced RNAs and for FL RNA nuclear export. CONCLUSIONS: By contrast to HIV-1, MLV recruits the same pathway for the cytoplasmic expression of its spliced and unspliced RNAs. Thus, MLV RNA expression depends upon coordinated splicing/export processes. In addition, FL RNA translation relies on Tap/NXF1-dependent export, raising the critical question of whether the pool of FL RNA to be packaged is also exported by Tap/NXF1.


Assuntos
Antígenos Ly/metabolismo , Expressão Gênica , Vírus da Leucemia Murina/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Western Blotting , Linhagem Celular , Citoplasma/metabolismo , Vírus da Leucemia Murina/genética , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais/biossíntese
17.
PLoS One ; 7(12): e51534, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23236513

RESUMO

Retroviral nucleocapsid proteins harbor nucleic acid chaperoning activities that mostly rely on the N-terminal basic residues and the CCHC zinc finger motif. Such chaperoning is essential for virus replication, notably for genomic RNA selection and packaging in virions, and for reverse transcription of genomic RNA into DNA. Recent data revealed that HIV-1 nucleocapsid restricts reverse transcription during virus assembly--a process called late reverse transcription--suggesting a regulation between RNA packaging and late reverse transcription. Indeed, mutating the HIV-1 nucleocapsid basic residues or the two zinc fingers caused a reduction in RNA incorporated and an increase in newly made viral DNA in the mutant virions. MoMuLV nucleocapsid has an N-terminal basic region similar to HIV-1 nucleocapsid but a unique zinc finger. This prompted us to investigate whether the N-terminal basic residues and the zinc finger of MoMuLV and HIV-1 nucleocapsids play a similar role in genomic RNA packaging and late reverse transcription. To this end, we analyzed the genomic RNA and viral DNA contents of virions produced by cells transfected with MoMuLV molecular clones where the zinc finger was mutated or completely deleted or with a deletion of the N-terminal basic residues of nucleocapsid. All mutant virions showed a strong defect in genomic RNA content indicating that the basic residues and zinc finger are important for genomic RNA packaging. In contrast to HIV-1 nucleocapsid-mutants, the level of viral DNA in mutant MoMuLV virions was only slightly increased. These results confirm that the N-terminal basic residues and zinc finger of MoMuLV nucleocapsid are critical for genomic RNA packaging but, in contrast to HIV-1 nucleocapsid, they most probably do not play a role in the control of late reverse transcription. In addition, these results suggest that virus formation and late reverse transcription proceed according to distinct mechanisms for MuLV and HIV-1.


Assuntos
HIV-1/fisiologia , Vírus da Leucemia Murina/fisiologia , Proteínas do Nucleocapsídeo/fisiologia , RNA Viral/fisiologia , Transcrição Reversa/fisiologia , Montagem de Vírus/fisiologia , Sequência de Aminoácidos , Células HEK293 , HIV-1/genética , Humanos , Vírus da Leucemia Murina/genética , Dados de Sequência Molecular , Mutagênese , Proteínas do Nucleocapsídeo/genética , Oligonucleotídeos/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Reversa/genética , Dedos de Zinco/genética , Dedos de Zinco/fisiologia
18.
Biochimie ; 94(12): 2498-507, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22766015

RESUMO

Treatment of HIV-1 with nucleoside reverse transcription inhibitors leads to the emergence of resistance mutations in the reverse transcriptase (RT) gene. Resistance to 3'-azido-3'-deoxythymidine (AZT) and to a lesser extent to 2'-3'-didehydro-2'-3'-dideoxythymidine is mediated by phosphorolytic excision of the chain terminator. Wild-type RT excises AZT by pyrophosphorolysis, while thymidine-associated resistance mutations in RT (TAMs) favour ATP as the donor substrate. However, in vitro, resistant RT still uses pyrophosphate more efficiently than ATP. We performed in vitro (-) strong-stop DNA synthesis experiments, with wild-type and AZT-resistant HIV-1 RTs, in the presence of physiologically relevant pyrophosphate and/or ATP concentrations and found that in the presence of pyrophosphate, ATP and AZTTP, TAMs do not enhance in vitro (-) strong-stop DNA synthesis. We hypothesized that utilisation of ATP in vivo is driven by intrinsic low pyrophosphate concentrations within the reverse transcription complex, which could be explained by the packaging of a cellular pyrophosphatase. We showed that over-expressed flagged-pyrophosphatase was associated with HIV-1 viral-like particles. In addition, we demonstrated that when HIV-1 particles were purified in order to avoid cellular microvesicle contamination, a pyrophosphatase activity was specifically associated to them. The presence of a pyrophosphatase activity in close proximity to the reverse transcription complex is most likely advantageous to the virus, even in the absence of any drug pressure.


Assuntos
Transcriptase Reversa do HIV/metabolismo , HIV-1/enzimologia , Pirofosfatases/metabolismo , Vírion/enzimologia , Trifosfato de Adenosina/metabolismo , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , DNA Viral/genética , DNA Viral/metabolismo , Didesoxinucleotídeos/metabolismo , Difosfatos/metabolismo , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Transcriptase Reversa do HIV/antagonistas & inibidores , Transcriptase Reversa do HIV/genética , HIV-1/efeitos dos fármacos , HIV-1/genética , Cinética , Mutação , Pirofosfatases/genética , Estavudina/metabolismo , Estavudina/farmacologia , Especificidade por Substrato , Nucleotídeos de Timina/metabolismo , Vírion/efeitos dos fármacos , Vírion/genética , Zidovudina/análogos & derivados , Zidovudina/metabolismo , Zidovudina/farmacologia
19.
RNA Biol ; 8(4): 572-80, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21691151

RESUMO

Generation of infectious retroviral particles rely on the targeting of all structural components to the correct cellular sites at the correct time. Gag, the main structural protein, orchestrates the assembly process and the mechanisms that trigger its targeting to assembly sites are well described. Gag is also responsible for the packaging of the viral genome and the molecular details of the Gag/RNA interaction are well characterized. Until recently, much less was understood about the cell biology of retrovirus RNA packaging. However, novel biochemical and live-cell microscopic approaches have identified where in the cell the initial events of genome recognition by Gag occur. These recent developments have shed light on the role played by the viral genome during virion assembly. Other central issues of the cell biology of RNA packaging, such as how the Gag-RNA complex traffics through the cytoplasm towards assembly sites, await characterization.


Assuntos
Produtos do Gene gag/metabolismo , Genoma Viral , RNA Viral/genética , RNA Viral/metabolismo , Retroviridae/fisiologia , Animais , Membrana Celular/virologia , Citoplasma/metabolismo , Produtos do Gene gag/genética , Humanos , Retroviridae/genética , Montagem de Vírus/genética , Montagem de Vírus/fisiologia
20.
RNA Biol ; 8(2): 343-53, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21422817

RESUMO

RNA-based compounds are promising agents to inactivate viruses. New specific hepatitis delta virus (HDV)-derived ribozymes are natural molecules that can be engineered to specifically target a viral RNA. We have designed specific on-off adaptor (SOFA)-HDV ribozymes targeting the tat and rev sequences of the human immunodeficiency virus type 1 (HIV-1) RNA. We show that the SOFA-HDV ribozymes cleave their RNA target in vitro. They inhibit the Tat-mediated transactivation of HIV-1 from 62% to 86% in different assays. In vivo, the amount of HIV RNA was decreased by 60 and 86% with two distinct ribozymes, which indicates that the inhibition of HIV production is directly correlated to the decline in spliced and unspliced viral RNAs. These SOFAHDV- ribozymes inhibited the expression and the viral production of four HIV-1 strains, indicating an extended potential to act on multiple HIV variants. In HEK 293T and HeLa cells transfected with pNL4-3 and the SOFA-HDV-ribozymes, the reduced RNA levels consequently decreased the Gag protein expression in the cell and virus production in the supernatant. When transfected before HIV-1 infection, the ribozymes prevented the incoming virus from being expressed. The ribozymes inhibited HIV production up to 90% when transfected in combination with the HIV protease inhibitor Atazanavir. Our results strongly suggest that SOFA-HDV ribozymes have a great potential to target HIV-1 and to be used as therapeutic agents in combination therapy.


Assuntos
HIV-1/enzimologia , RNA Catalítico/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Replicação Viral , Sulfato de Atazanavir , Sequência de Bases , Produtos do Gene gag/biossíntese , Produtos do Gene gag/genética , Produtos do Gene rev/genética , Produtos do Gene rev/metabolismo , Células HEK293 , Infecções por HIV/genética , HIV-1/genética , Células HeLa , Vírus Delta da Hepatite/enzimologia , Vírus Delta da Hepatite/genética , Humanos , Oligopeptídeos/farmacologia , Piridinas/farmacologia , Splicing de RNA , RNA Catalítico/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA