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












Base de datos
Intervalo de año de publicación
1.
Retrovirology ; 20(1): 16, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37700325

RESUMEN

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.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Animales , Genómica , Virus de la Leucemia Murina/genética , Regiones Promotoras Genéticas , ARN
2.
Biophys Rep (N Y) ; 2(3): 100068, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36425325

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-35475610

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares , Nanopartículas , Nanoporos , Virus , Virión
4.
J Biol Chem ; 298(1): 101500, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34929171

RESUMEN

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.


Asunto(s)
Productos del Gen gag , VIH-1 , Nucleoproteínas , Regiones no Traducidas 5' , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Genómica , VIH-1/genética , VIH-1/metabolismo , Microscopía Electrónica de Transmisión , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , ARN Guía de Kinetoplastida , ARN Viral/genética , ARN Viral/metabolismo , Ensamble de Virus/genética
5.
Viruses ; 12(9)2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32899657

RESUMEN

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.


Asunto(s)
Infecciones por VIH/diagnóstico , VIH-1/fisiología , Técnicas Analíticas Microfluídicas , Microfluídica , Síndrome de Inmunodeficiencia Adquirida/diagnóstico , Síndrome de Inmunodeficiencia Adquirida/fisiopatología , Progresión de la Enfermedad , Infecciones por VIH/virología , VIH-1/genética , Humanos , Replicación Viral
6.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641479

RESUMEN

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.


Asunto(s)
Citidina Desaminasa/genética , Regulación Viral de la Expresión Génica , Productos del Gen gag/genética , Leucemia Experimental/genética , Virus de la Leucemia Murina de Moloney/genética , Infecciones por Retroviridae/genética , Infecciones Tumorales por Virus/genética , Empalme Alternativo , Animales , Cápside/metabolismo , Citidina Desaminasa/deficiencia , Productos del Gen gag/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/virología , Ratones , Ratones Noqueados , Virus de la Leucemia Murina de Moloney/metabolismo , Virus de la Leucemia Murina de Moloney/patogenicidad , Células 3T3 NIH , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Transducción de Señal , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/virología , Virión/genética , Virión/metabolismo , Virión/patogenicidad , Replicación Viral
7.
Nanoscale ; 11(13): 6036-6044, 2019 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-30869094

RESUMEN

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.


Asunto(s)
VIH-1/fisiología , Tetraspanina 28/química , Tetraspanina 29/química , Membrana Celular/metabolismo , Citometría de Flujo , Células HeLa , Humanos , Microscopía de Fuerza Atómica , Tetraspanina 28/metabolismo , Tetraspanina 29/metabolismo , Ensamble de Virus , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
8.
Nucleic Acids Res ; 46(9): e57, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29514260

RESUMEN

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.


Asunto(s)
VIH-1/genética , ARN Viral/biosíntesis , ARN Viral/química , Secuencias Reguladoras de Ácido Ribonucleico , Ensamble de Virus , Regiones no Traducidas 5' , Genoma Viral , Células HEK293 , VIH-1/fisiología , Humanos , Mutación , Motivos de Nucleótidos , Poli A/metabolismo , Replicación Viral
9.
Viruses ; 9(10)2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28961190

RESUMEN

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.


Asunto(s)
Terapia Genética , Infecciones por VIH/terapia , VIH-1/efectos de los fármacos , Inmunoterapia Adoptiva , Péptidos/uso terapéutico , Proteínas/uso terapéutico , Anticuerpos de Cadena Única/uso terapéutico , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/inmunología , Repetición de Anquirina , Anticuerpos Neutralizantes/uso terapéutico , Terapia Antirretroviral Altamente Activa/efectos adversos , Farmacorresistencia Viral , Anticuerpos Anti-VIH/inmunología , Anticuerpos Anti-VIH/uso terapéutico , Infecciones por VIH/inmunología , VIH-1/inmunología , Humanos , Integración Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
10.
J Virol ; 91(15)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28490595

RESUMEN

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.


Asunto(s)
VIH-1/inmunología , VIH-1/fisiología , Inmunidad Innata , Interferón Tipo I/metabolismo , Macrófagos/inmunología , Macrófagos/virología , Internalización del Virus , Células Cultivadas , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Tiempo
11.
Viruses ; 8(8)2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27556480

RESUMEN

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.


Asunto(s)
Biología Molecular/métodos , ARN Viral/metabolismo , Retroviridae/fisiología , Virión/metabolismo , Virología/métodos , Ensamble de Virus , Animales , Humanos
12.
Nucleic Acids Res ; 44(16): 7922-34, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27280976

RESUMEN

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.


Asunto(s)
Dimerización , VIH-1/genética , Imagenología Tridimensional , Microscopía/métodos , Conformación de Ácido Nucleico , ARN Viral/química , Membrana Celular/metabolismo , Supervivencia Celular , Color , Citosol/metabolismo , Genoma Viral , Células HeLa , Humanos , Hibridación Fluorescente in Situ , ARN Viral/genética , Coloración y Etiquetado , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
13.
Sci Rep ; 6: 27536, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27273064

RESUMEN

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.


Asunto(s)
VIH-1/genética , Interacciones Huésped-Patógeno , Nucleocápside/genética , Transcripción Reversa , Ensamble de Virus/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Desaminasa APOBEC-3G/genética , Desaminasa APOBEC-3G/metabolismo , Secuencia de Bases , Regulación de la Expresión Génica , Células HEK293 , VIH-1/metabolismo , Humanos , Secuencias Invertidas Repetidas , Nucleocápside/metabolismo , Unión Proteica , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Eliminación de Secuencia , Transducción de Señal , Virión/genética , Virión/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
14.
RNA Biol ; 12(9): 942-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26158194

RESUMEN

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.


Asunto(s)
Transporte Activo de Núcleo Celular , Intrones , Virus de la Leucemia Murina/fisiología , ARN Viral/genética , ARN Viral/metabolismo , Empalme Alternativo , Animales , Genoma Viral , Humanos , Secuencias Invertidas Repetidas , Ratones , Conformación de Ácido Nucleico , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Motivos de Nucleótidos , Sistemas de Lectura Abierta , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/química , Secuencias Reguladoras de Ácido Ribonucleico , Transducción de Señal , Secuencias Repetidas Terminales
15.
Nucleic Acids Res ; 43(1): 336-47, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25488808

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , VIH-1/fisiología , Factores de Transcripción/metabolismo , Ensamble de Virus , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Membrana Celular/virología , ADN Viral/biosíntesis , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Humanos , Transcripción Reversa , Eliminación de Secuencia , Virión/metabolismo , Dedos de Zinc , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química
16.
Retrovirology ; 11: 21, 2014 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-24597485

RESUMEN

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.


Asunto(s)
Antígenos Ly/metabolismo , Expresión Génica , Virus de la Leucemia Murina/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Transporte Activo de Núcleo Celular , Animales , Western Blotting , Línea Celular , Citoplasma/metabolismo , Virus de la Leucemia Murina/genética , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Virales/biosíntesis
17.
PLoS One ; 7(12): e51534, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23236513

RESUMEN

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.


Asunto(s)
VIH-1/fisiología , Virus de la Leucemia Murina/fisiología , Proteínas de la Nucleocápside/fisiología , ARN Viral/fisiología , Transcripción Reversa/fisiología , Ensamble de Virus/fisiología , Secuencia de Aminoácidos , Células HEK293 , VIH-1/genética , Humanos , Virus de la Leucemia Murina/genética , Datos de Secuencia Molecular , Mutagénesis , Proteínas de la Nucleocápside/genética , Oligonucleótidos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Reversa/genética , Dedos de Zinc/genética , Dedos de Zinc/fisiología
18.
Biochimie ; 94(12): 2498-507, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22766015

RESUMEN

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.


Asunto(s)
Transcriptasa Inversa del VIH/metabolismo , VIH-1/enzimología , Pirofosfatasas/metabolismo , Virión/enzimología , Adenosina Trifosfato/metabolismo , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/farmacología , ADN Viral/genética , ADN Viral/metabolismo , Didesoxinucleótidos/metabolismo , Difosfatos/metabolismo , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Transcriptasa Inversa del VIH/genética , VIH-1/efectos de los fármacos , VIH-1/genética , Cinética , Mutación , Pirofosfatasas/genética , Estavudina/metabolismo , Estavudina/farmacología , Especificidad por Sustrato , Nucleótidos de Timina/metabolismo , Virión/efectos de los fármacos , Virión/genética , Zidovudina/análogos & derivados , Zidovudina/metabolismo , Zidovudina/farmacología
19.
RNA Biol ; 8(4): 572-80, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21691151

RESUMEN

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.


Asunto(s)
Productos del Gen gag/metabolismo , Genoma Viral , ARN Viral/genética , ARN Viral/metabolismo , Retroviridae/fisiología , Animales , Membrana Celular/virología , Citoplasma/metabolismo , Productos del Gen gag/genética , Humanos , Retroviridae/genética , Ensamble de Virus/genética , Ensamble de Virus/fisiología
20.
RNA Biol ; 8(2): 343-53, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21422817

RESUMEN

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.


Asunto(s)
VIH-1/enzimología , ARN Catalítico/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Replicación Viral , Sulfato de Atazanavir , Secuencia de Bases , Productos del Gen gag/biosíntesis , Productos del Gen gag/genética , Productos del Gen rev/genética , Productos del Gen rev/metabolismo , Células HEK293 , Infecciones por VIH/genética , VIH-1/genética , Células HeLa , Virus de la Hepatitis Delta/enzimología , Virus de la Hepatitis Delta/genética , Humanos , Oligopéptidos/farmacología , Piridinas/farmacología , Empalme del ARN , ARN Catalítico/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...