Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nat Rev Genet ; 18(5): 292-308, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28286338

RESUMEN

Transposable elements and retroviruses are found in most genomes, can be pathogenic and are widely used as gene-delivery and functional genomics tools. Exploring whether these genetic elements target specific genomic sites for integration and how this preference is achieved is crucial to our understanding of genome evolution, somatic genome plasticity in cancer and ageing, host-parasite interactions and genome engineering applications. High-throughput profiling of integration sites by next-generation sequencing, combined with large-scale genomic data mining and cellular or biochemical approaches, has revealed that the insertions are usually non-random. The DNA sequence, chromatin and nuclear context, and cellular proteins cooperate in guiding integration in eukaryotic genomes, leading to a remarkable diversity of insertion site distribution and evolutionary strategies.


Asunto(s)
Elementos Transponibles de ADN/genética , Eucariontes/genética , Variación Genética/genética , Genoma Viral , Genómica/métodos , Retroviridae/genética , Integración Viral/genética , Animales , Evolución Molecular , Humanos
2.
Retrovirology ; 19(1): 23, 2022 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-36309692

RESUMEN

BACKGROUND: TASOR, a component of the HUSH repressor epigenetic complex, and SAMHD1, a cellular triphosphohydrolase (dNTPase), are both anti-HIV proteins antagonized by HIV-2/SIVsmm Viral protein X. As a result, the same viral protein is able to relieve two different blocks along the viral life cell cycle, one at the level of reverse transcription, by degrading SAMHD1, the other one at the level of proviral expression, by degrading TASOR. Phosphorylation of SAMHD1 at T592 has been shown to downregulate its antiviral activity. The discovery that T819 in TASOR was lying within a SAMHD1 T592-like motif led us to ask whether TASOR is phosphorylated on this residue and whether this post-translational modification could regulate its repressive activity. RESULTS: Using a specific anti-phospho-antibody, we found that TASOR is phosphorylated at T819, especially in cells arrested in early mitosis by nocodazole. We provide evidence that the phosphorylation is conducted by a Cyclin/CDK1 complex, like that of SAMHD1 at T592. While we could not detect TASOR in quiescent CD4 + T cells, TASOR and its phosphorylated form are present in activated primary CD4 + T lymphocytes. In addition, TASOR phosphorylation appears to be independent from TASOR repressive activity. Indeed, on the one hand, nocodazole barely reactivates HIV-1 in the J-Lat A1 HIV-1 latency model despite TASOR T819 phosphorylation. On the other hand, etoposide, a second cell cycle arresting drug, reactivates latent HIV-1, without concomitant TASOR phosphorylation. Furthermore, overexpression of wt TASOR or T819A or T819E similarly represses gene expression driven by an HIV-1-derived LTR promoter. Finally, while TASOR is degraded by HIV-2 Vpx, TASOR phosphorylation is prevented by HIV-1 Vpr, likely as a consequence of HIV-1 Vpr-mediated-G2 arrest. CONCLUSIONS: Altogether, we show that TASOR phosphorylation occurs in vivo on T819. This event does not appear to correlate with TASOR-mediated HIV-1 silencing. We speculate that TASOR phosphorylation is related to a role of TASOR during cell cycle progression.


Asunto(s)
Infecciones por VIH , VIH-1 , Proteínas de Unión al GTP Monoméricas , Humanos , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , VIH-1/fisiología , Fosforilación , Treonina , Nocodazol/metabolismo , Latencia del Virus , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Nucleares/metabolismo
3.
J Virol ; 94(7)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-31915280

RESUMEN

Dengue virus (DENV) is a mosquito-borne flavivirus responsible for dengue disease, a major human health concern for which no specific therapies are available. Like other viruses, DENV relies heavily on the host cellular machinery for productive infection. In this study, we performed a genome-wide CRISPR-Cas9 screen using haploid HAP1 cells to identify host genes important for DENV infection. We identified DPM1 and -3, two subunits of the endoplasmic reticulum (ER) resident dolichol-phosphate mannose synthase (DPMS) complex, as host dependency factors for DENV and other related flaviviruses, such as Zika virus (ZIKV). The DPMS complex catalyzes the synthesis of dolichol-phosphate mannose (DPM), which serves as mannosyl donor in pathways leading to N-glycosylation, glycosylphosphatidylinositol (GPI) anchor biosynthesis, and C- or O-mannosylation of proteins in the ER lumen. Mutation in the DXD motif of DPM1, which is essential for its catalytic activity, abolished DPMS-mediated DENV infection. Similarly, genetic ablation of ALG3, a mannosyltransferase that transfers mannose to lipid-linked oligosaccharide (LLO), rendered cells poorly susceptible to DENV. We also established that in cells deficient for DPMS activity, viral RNA amplification is hampered and truncated oligosaccharides are transferred to the viral prM and E glycoproteins, affecting their proper folding. Overall, our study provides new insights into the host-dependent mechanisms of DENV infection and supports current therapeutic approaches using glycosylation inhibitors to treat DENV infection.IMPORTANCE Dengue disease, which is caused by dengue virus (DENV), has emerged as the most important mosquito-borne viral disease in humans and is a major global health concern. DENV encodes only few proteins and relies on the host cell machinery to accomplish its life cycle. The identification of the host factors important for DENV infection is needed to propose new targets for antiviral intervention. Using a genome-wide CRISPR-Cas9 screen, we identified DPM1 and -3, two subunits of the DPMS complex, as important host factors for the replication of DENV as well as other related viruses such as Zika virus. We established that DPMS complex plays dual roles during viral infection, both regulating viral RNA replication and promoting viral structural glycoprotein folding/stability. These results provide insights into the host molecules exploited by DENV and other flaviviruses to facilitate their life cycle.


Asunto(s)
Sistemas CRISPR-Cas , Virus del Dengue/fisiología , Dengue/virología , Manosiltransferasas/metabolismo , Animales , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Fibroblastos/metabolismo , Glicoproteínas/metabolismo , Glicosilación , Glicosilfosfatidilinositoles/metabolismo , Células HEK293 , Humanos , Manosa/química , Oligosacáridos/química , ARN Guía de Kinetoplastida/metabolismo , ARN Viral/química , Células Vero , Replicación Viral
4.
J Biol Chem ; 293(16): 6172-6186, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29507092

RESUMEN

Recently, a new class of HIV-1 integrase (IN) inhibitors with a dual mode of action, called IN-LEDGF/p75 allosteric inhibitors (INLAIs), was described. Designed to interfere with the IN-LEDGF/p75 interaction during viral integration, unexpectedly, their major impact was on virus maturation. This activity has been linked to induction of aberrant IN multimerization, whereas inhibition of the IN-LEDGF/p75 interaction accounts for weaker antiretroviral effect at integration. Because these dual activities result from INLAI binding to IN at a single binding site, we expected that these activities co-evolved together, driven by the affinity for IN. Using an original INLAI, MUT-A, and its activity on an Ala-125 (A125) IN variant, we found that these two activities on A125-IN can be fully dissociated: MUT-A-induced IN multimerization and the formation of eccentric condensates in viral particles, which are responsible for inhibition of virus maturation, were lost, whereas inhibition of the IN-LEDGF/p75 interaction and consequently integration was fully retained. Hence, the mere binding of INLAI to A125 IN is insufficient to promote the conformational changes of IN required for aberrant multimerization. By analyzing the X-ray structures of MUT-A bound to the IN catalytic core domain (CCD) with or without the Ala-125 polymorphism, we discovered that the loss of IN multimerization is due to stabilization of the A125-IN variant CCD dimer, highlighting the importance of the CCD dimerization energy for IN multimerization. Our study reveals that affinity for the LEDGF/p75-binding pocket is not sufficient to induce INLAI-dependent IN multimerization and the associated inhibition of viral maturation.


Asunto(s)
Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/efectos de los fármacos , VIH-1/fisiología , Ensamble de Virus/efectos de los fármacos , Integración Viral/efectos de los fármacos , Regulación Alostérica , Sitios de Unión , Línea Celular , Inhibidores de Integrasa VIH/química , Humanos , Estructura Molecular , Piridinas/química , Piridinas/farmacología , Relación Estructura-Actividad , Tiofenos/química , Tiofenos/farmacología
5.
Retrovirology ; 15(1): 48, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29996845

RESUMEN

BACKGROUND: Nuclear localization of Gag is a property shared by many retroviruses and retrotransposons. The importance of this stage for retroviral replication is still unknown, but studies on the Rous Sarcoma virus indicate that Gag might select the viral RNA genome for packaging in the nucleus. In the case of Foamy viruses, genome encapsidation is mediated by Gag C-terminal domain (CTD), which harbors three clusters of glycine and arginine residues named GR boxes (GRI-III). In this study we investigated how PFV Gag subnuclear distribution might be regulated. RESULTS: We show that the isolated GRI and GRIII boxes act as nucleolar localization signals. In contrast, both the entire Gag CTD and the isolated GRII box, which contains the chromatin-binding motif, target the nucleolus exclusively upon mutation of the evolutionary conserved arginine residue at position 540 (R540), which is a key determinant of FV Gag chromatin tethering. We also provide evidence that Gag localizes in the nucleolus during FV replication and uncovered that the viral protein interacts with and is methylated by Protein Arginine Methyltransferase 1 (PRMT1) in a manner that depends on the R540 residue. Finally, we show that PRMT1 depletion by RNA interference induces the concentration of Gag C-terminus in nucleoli. CONCLUSION: Altogether, our findings suggest that methylation by PRMT1 might finely tune the subnuclear distribution of Gag depending on the stage of the FV replication cycle. The role of this step for viral replication remains an open question.


Asunto(s)
Secuencias de Aminoácidos , Arginina , Productos del Gen gag/metabolismo , Dominios y Motivos de Interacción de Proteínas , Infecciones por Retroviridae/virología , Spumavirus/fisiología , Secuencia de Aminoácidos , Arginina/química , Núcleo Celular/metabolismo , Cromatina/metabolismo , Evolución Molecular , Productos del Gen gag/química , Productos del Gen gag/genética , Humanos , Señales de Localización Nuclear , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Infecciones por Retroviridae/metabolismo
6.
Retrovirology ; 14(1): 50, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-29121950

RESUMEN

BACKGROUND: HIV-1 Integrase (IN) interacts with the cellular co-factor LEDGF/p75 and tethers the HIV preintegration complex to the host genome enabling integration. Recently a new class of IN inhibitors was described, the IN-LEDGF allosteric inhibitors (INLAIs). Designed to interfere with the IN-LEDGF interaction during integration, the major impact of these inhibitors was surprisingly found on virus maturation, causing a reverse transcription defect in target cells. RESULTS: Here we describe the MUT-A compound as a genuine INLAI with an original chemical structure based on a new type of scaffold, a thiophene ring. MUT-A has all characteristics of INLAI compounds such as inhibition of IN-LEDGF/p75 interaction, IN multimerization, dual antiretroviral (ARV) activities, normal packaging of genomic viral RNA and complete Gag protein maturation. MUT-A has more potent ARV activity compared to other INLAIs previously reported, but similar profile of resistance mutations and absence of ARV activity on SIV. HIV-1 virions produced in the presence of MUT-A were non-infectious with the formation of eccentric condensates outside of the core. In studying the immunoreactivity of these non-infectious virions, we found that inactivated HIV-1 particles were captured by anti-HIV-specific neutralizing and non-neutralizing antibodies (b12, 2G12, PGT121, 4D4, 10-1074, 10E8, VRC01) with efficiencies comparable to non-treated virus. Autologous CD4+ T lymphocyte proliferation and cytokine induction by monocyte-derived dendritic cells (MDDC) pulsed either with MUT-A-inactivated HIV or non-treated HIV were also comparable. CONCLUSIONS: Although strongly defective in infectivity, HIV-1 virions produced in the presence of the MUT-A INLAI have a normal protein and genomic RNA content as well as B and T cell immunoreactivities comparable to non-treated HIV-1. These inactivated viruses might form an attractive new approach in vaccine research in an attempt to study if this new type of immunogen could elicit an immune response against HIV-1 in animal models.


Asunto(s)
Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/metabolismo , VIH-1/efectos de los fármacos , VIH-1/enzimología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Piridinas/farmacología , Tiofenos/farmacología , Línea Celular , Anticuerpos Anti-VIH/inmunología , Inhibidores de Integrasa VIH/química , VIH-1/inmunología , Humanos , Piridinas/química , Tiofenos/química , Ensamble de Virus/efectos de los fármacos , Integración Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
Bioinformatics ; 31(21): 3483-91, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26142185

RESUMEN

MOTIVATION: Post-translational modification by the Small Ubiquitin-like Modifier (SUMO) proteins, a process termed SUMOylation, is involved in many fundamental cellular processes. SUMO proteins are conjugated to a protein substrate, creating an interface for the recruitment of cofactors harboring SUMO-interacting motifs (SIMs). Mapping both SUMO-conjugation sites and SIMs is required to study the functional consequence of SUMOylation. To define the best candidate sites for experimental validation we designed JASSA, a Joint Analyzer of SUMOylation site and SIMs. RESULTS: JASSA is a predictor that uses a scoring system based on a Position Frequency Matrix derived from the alignment of experimental SUMOylation sites or SIMs. Compared with existing web-tools, JASSA displays on par or better performances. Novel features were implemented towards a better evaluation of the prediction, including identification of database hits matching the query sequence and representation of candidate sites within the secondary structural elements and/or the 3D fold of the protein of interest, retrievable from deposited PDB files. AVAILABILITY AND IMPLEMENTATION: JASSA is freely accessible at http://www.jassa.fr/. Website is implemented in PHP and MySQL, with all major browsers supported. CONTACT: guillaume.beauclair@inserm.fr SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Análisis de Secuencia de Proteína/métodos , Programas Informáticos , Sumoilación , Humanos , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Alineación de Secuencia , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo
8.
Retrovirology ; 12: 24, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25808736

RESUMEN

BACKGROUND: Genomic integration, an obligate step in the HIV-1 replication cycle, is blocked by the integrase inhibitor raltegravir. A consequence is an excess of unintegrated viral DNA genomes, which undergo intramolecular ligation and accumulate as 2-LTR circles. These circularized genomes are also reliably observed in vivo in the absence of antiviral therapy and they persist in non-dividing cells. However, they have long been considered as dead-end products that are not precursors to integration and further viral propagation. RESULTS: Here, we show that raltegravir action is reversible and that unintegrated viral DNA is integrated in the host cell genome after raltegravir removal leading to HIV-1 replication. Using quantitative PCR approach, we analyzed the consequences of reversing prolonged raltegravir-induced integration blocks. We observed, after RAL removal, a decrease of 2-LTR circles and a transient increase of linear DNA that is subsequently integrated in the host cell genome and fuel new cycles of viral replication. CONCLUSIONS: Our data highly suggest that 2-LTR circles can be used as a reserve supply of genomes for proviral integration highlighting their potential role in the overall HIV-1 replication cycle.


Asunto(s)
ADN Viral/metabolismo , Integrasa de VIH/metabolismo , VIH-1/fisiología , Integración Viral , Replicación Viral , Línea Celular , Inhibidores de Integrasa VIH/metabolismo , VIH-1/enzimología , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
J Virol ; 87(7): 3729-40, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23325686

RESUMEN

Nef is a human immunodeficiency virus type 1 (HIV-1) auxiliary protein that plays an important role in virus replication and the onset of acquired immunodeficiency. Although known functions of Nef might explain its contribution to HIV-1-associated pathogenesis, how Nef increases virus infectivity is still an open question. In vitro, Nef-deleted viruses have a defect that prevents efficient completion of early steps of replication. We have previously shown that this restriction is not due to the absence of Nef in viral particles. Rather, a loss of function in virus-producing cells accounts for the lower infectivity of nef-deleted viruses compared to wild-type (WT) viruses. Here we used DiGE and iTRAQ to identify differences between the proteomes of WT and nef-deleted viruses. We observe that glucosidase II is enriched in WT virions, whereas Ezrin, ALG-2, CD81, and EHD4 are enriched in nef-deleted virions. Functional analysis shows that glucosidase II, ALG-2, and CD81 have no or only Nef-independent effect on infectivity. In contrast, Ezrin and EHD4 are involved in the ability of Nef to increase virus infectivity (referred to thereafter as Nef potency). Indeed, simultaneous Ezrin and EHD4 depletion in SupT1 and 293T virus-producing cells result in an ∼30 and ∼70% decrease of Nef potency, respectively. Finally, while Ezrin behaves as an inhibitory factor counteracted by Nef, EHD4 should be considered as a cofactors required by Nef to increase virus infectivity.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , VIH-1/genética , VIH-1/patogenicidad , Proteínas Nucleares/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/deficiencia , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Western Blotting , Electroforesis/métodos , Silenciador del Gen , Células HEK293 , VIH-1/metabolismo , Humanos , Proteómica , ARN Interferente Pequeño , Tetraspanina 28/metabolismo , Ultracentrifugación , Virión/metabolismo , alfa-Glucosidasas/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
10.
Retrovirology ; 10: 144, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24261564

RESUMEN

BACKGROUND: LEDGF/p75 (LEDGF) is the main cellular cofactor of HIV-1 integrase (IN). It acts as a tethering factor for IN, and targets the integration of HIV in actively transcribed gene regions of chromatin. A recently developed class of IN allosteric inhibitors can inhibit the LEDGF-IN interaction. RESULTS: We describe a new series of IN-LEDGF allosteric inhibitors, the most active of which is Mut101. We determined the crystal structure of Mut101 in complex with IN and showed that the compound binds to the LEDGF-binding pocket, promoting conformational changes of IN which explain at the atomic level the allosteric effect of the IN/LEDGF interaction inhibitor on IN functions. In vitro, Mut101 inhibited both IN-LEDGF interaction and IN strand transfer activity while enhancing IN-IN interaction. Time of addition experiments indicated that Mut101 behaved as an integration inhibitor. Mut101 was fully active on HIV-1 mutants resistant to INSTIs and other classes of anti-HIV drugs, indicative that this compound has a new mode of action. However, we found that Mut101 also displayed a more potent antiretroviral activity at a post-integration step. Infectivity of viral particles produced in presence of Mut101 was severely decreased. This latter effect also required the binding of the compound to the LEDGF-binding pocket. CONCLUSION: Mut101 has dual anti-HIV-1 activity, at integration and post-integration steps of the viral replication cycle, by binding to a unique target on IN (the LEDGF-binding pocket). The post-integration block of HIV-1 replication in virus-producer cells is the mechanism by which Mut101 is most active as an antiretroviral. To explain this difference between Mut101 antiretroviral activity at integration and post-integration stages, we propose the following model: LEDGF is a nuclear, chromatin-bound protein that is absent in the cytoplasm. Therefore, LEDGF can outcompete compound binding to IN in the nucleus of target cells lowering its antiretroviral activity at integration, but not in the cytoplasm where post-integration production of infectious viral particles takes place.


Asunto(s)
Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/metabolismo , VIH-1/efectos de los fármacos , VIH-1/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Integración Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Línea Celular , Cristalografía por Rayos X , Integrasa de VIH/química , Inhibidores de Integrasa VIH/química , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Unión Proteica , Conformación Proteica
11.
Blood ; 117(1): 190-9, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20959607

RESUMEN

The human T-lymphotropic virus type I oncoprotein Tax is critical for T-cell transformation, acting mainly through nuclear factor kappa B essential modulator (NEMO) binding and subsequent nuclear factor-κB activation. Tax localizes to Tax nuclear bodies and to the centrosome and is subjected to ubiquitylation and small ubiquitin-like modifier (SUMO)ylation, which are both necessary for complete transcriptional activation. Using the photoconvertible fluorophore Dendra-2 coupled with live video confocal microscopy, we show for the first time that the same Tax molecule shuttles among Tax nuclear bodies and between these nuclear bodies and the centrosome, depending on its posttranslational modifications. Ubiquitylation targets Tax to nuclear bodies to which NEMO is recruited and subsequently SUMOylated. We also demonstrate that Tax nuclear bodies contain the SUMOylation machinery including SUMO and the SUMO conjugating enzyme Ubc9, strongly suggesting that these nuclear bodies represent sites of active SUMOylation. Finally, both ubiquitylation and SUMOylation of Tax control NEMO targeting to the centrosome. Altogether, we are proposing a model where both ubiquitylation and SUMOylation of Tax control the shuttling of Tax and NEMO between the cytoplasmic and nuclear compartments.


Asunto(s)
Núcleo Celular/metabolismo , Centrosoma/fisiología , Productos del Gen tax/fisiología , Quinasa I-kappa B/metabolismo , Sumoilación , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación , Western Blotting , Células Cultivadas , Citoplasma/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Quinasa I-kappa B/genética , Riñón/citología , Riñón/metabolismo , Pulmón/citología , Pulmón/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Activación Transcripcional , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética
12.
J Biol Chem ; 286(23): 21013-22, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21454548

RESUMEN

HIV-1 integrase (IN) orchestrates the integration of the reverse transcribed viral cDNA into the host cell genome and participates also in other steps of HIV-1 replication. Cellular and viral factors assist IN in performing its multiple functions, and post-translational modifications contribute to modulate its activities. Here, we show that HIV-1 IN is modified by SUMO proteins and that phylogenetically conserved SUMOylation consensus motifs represent major SUMO acceptor sites. Viruses harboring SUMOylation site IN mutants displayed a replication defect that was mapped during the early stages of infection, before integration but after reverse transcription. Because SUMOylation-defective IN mutants retained WT catalytic activity, we hypothesize that SUMOylation might regulate the affinity of IN for co-factors, contributing to efficient HIV-1 replication.


Asunto(s)
Infecciones por VIH/metabolismo , Integrasa de VIH/metabolismo , VIH-1/fisiología , Sumoilación/fisiología , Integración Viral/fisiología , Replicación Viral/fisiología , Células HEK293 , Infecciones por VIH/genética , Integrasa de VIH/genética , Células HeLa , Humanos , Mutación , Transcripción Reversa/fisiología
13.
Cell Mol Life Sci ; 68(20): 3385-99, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21337044

RESUMEN

Vγ9Vδ2 T cells play a crucial role in the antitumoral immune response through cytokine production and cytotoxicity. Although the expression of the immunomodulatory molecule HLA-G has been found in diverse tumors, its impact on Vγ9Vδ2 T-cell functions remains unknown. Here we showed that soluble HLA-G inhibits Vγ9Vδ2 T-cell proliferation without inducing apoptosis. Moreover, soluble HLA-G inhibited the Vγ9Vδ2 T-cell production of IFN-γ induced by phosphoantigen stimulation. The reduction in Vγ9Vδ2 T-cell IFN-γ production was also induced by membrane-bound or soluble HLA-G expressed by tumor cell lines. Finally, primary tumor cells inhibited Vγ9Vδ2 T-cell proliferation and IFN-γ production through HLA-G. In this context, HLA-G impaired Vγ9Vδ2 T-cell cytotoxicity by interacting with ILT2 inhibitory receptor. These data demonstrate that HLA-G inhibits the anti-tumoral functions of Vγ9Vδ2 T cells and imply that treatments targeting HLA-G could optimize Vγ9Vδ2 T-cell-mediated immunotherapy of cancer.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoterapia , Activación de Linfocitos/inmunología , Melanoma/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/antagonistas & inhibidores , Linfocitos T/inmunología , Antígenos CD/metabolismo , Apoptosis , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Antígenos HLA-G , Humanos , Interferón gamma/metabolismo , Receptor Leucocitario Tipo Inmunoglobulina B1 , Melanoma/metabolismo , Melanoma/terapia , Glicoproteínas de Membrana/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores Inmunológicos/metabolismo , Receptores KIR2DL4/metabolismo , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología
14.
Virologie (Montrouge) ; 16(2): 73-84, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-31881589

RESUMEN

Integration into the genome of the host cell is an obligatory step in the replication of retroelements. This feature accounts for the fact that these elements are both potential mutagens as well as vectors suitable for long-term gene therapy. Recently, many studies have reported that proviral insertion is not random but, rather, targets specific regions in the genome. Additionally, it has become clear that this process is highly regulated at the molecular level. Both viral proteins and cellular factors participate in the integration step, explaining why different retroelements have distinct integration profiles. This review describes recent advances about the integration of retroelements, focusing particularly on the mechanisms involved in the selectivity and specificity of integration and the chromatin-anchoring step, which precedes the insertion of the provirus.

15.
Biochim Biophys Acta ; 1799(3-4): 207-16, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19683084

RESUMEN

Since integration into the host cell genome is an obligatory step for their replication, retro-elements are both potent insertional mutagens and also suitable vectors for gene therapy. Many recent studies reported that the integration process is not random but, on the contrary, higly regulated at the molecular level. Many viral proteins and cellular factors play a key role in the integration step, explaining the reason why different retro-elements display distinct integration profiles. This review describes the recent highlights about integration of retro-elements with particular focus on the mechanisms underlying the specificity of integration target-site selection and the step of chromosomal tethering which preceeds insertion of the provirus.


Asunto(s)
Cromosomas Humanos/genética , Integración Viral/fisiología , VIH-1/fisiología , Humanos , Lentivirus/fisiología , Retroelementos/fisiología , Replicación Viral/fisiología
16.
Dev Cell ; 10(6): 821-30, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16740483

RESUMEN

The vacuolar protein sorting machinery regulates multivesicular body biogenesis and is selectively recruited by enveloped viruses to support budding. Here we report the crystal structure of the human ESCRT-III protein CHMP3 at 2.8 A resolution. The core structure of CHMP3 folds into a flat helical arrangement that assembles into a lattice, mainly via two different dimerization modes, and unilaterally exposes a highly basic surface. The C terminus, the target for Vps4-induced ESCRT disassembly, extends from the opposite side of the membrane targeting region. Mutations within the basic and dimerization regions hinder bilayer interaction in vivo and reverse the dominant-negative effect of a truncated CHMP3 fusion protein on HIV-1 budding. Thus, the final steps in the budding process may include CHMP protein polymerization and lattice formation on membranes by employing different bilayer-recognizing surfaces, a function shared by all CHMP family members.


Asunto(s)
Cristalografía por Rayos X , Infecciones por VIH , VIH-1/fisiología , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Aminoácidos Acídicos/química , Aminoácidos Básicos/química , Secuencia Conservada , Dimerización , Complejos de Clasificación Endosomal Requeridos para el Transporte , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Estructura Secundaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Electricidad Estática , Proteínas de Transporte Vesicular/genética , Replicación Viral
17.
Nat Commun ; 12(1): 4582, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321470

RESUMEN

SAMHD1 is a cellular triphosphohydrolase (dNTPase) proposed to inhibit HIV-1 reverse transcription in non-cycling immune cells by limiting the supply of the dNTP substrates. Yet, phosphorylation of T592 downregulates SAMHD1 antiviral activity, but not its dNTPase function, implying that additional mechanisms contribute to viral restriction. Here, we show that SAMHD1 is SUMOylated on residue K595, a modification that relies on the presence of a proximal SUMO-interacting motif (SIM). Loss of K595 SUMOylation suppresses the restriction activity of SAMHD1, even in the context of the constitutively active phospho-ablative T592A mutant but has no impact on dNTP depletion. Conversely, the artificial fusion of SUMO2 to a non-SUMOylatable inactive SAMHD1 variant restores its antiviral function, a phenotype that is reversed by the phosphomimetic T592E mutation. Collectively, our observations clearly establish that lack of T592 phosphorylation cannot fully account for the restriction activity of SAMHD1. We find that SUMOylation of K595 is required to stimulate a dNTPase-independent antiviral activity in non-cycling immune cells, an effect that is antagonized by cyclin/CDK-dependent phosphorylation of T592 in cycling cells.


Asunto(s)
Ciclo Celular/fisiología , VIH-1/fisiología , Proteína 1 que Contiene Dominios SAM y HD/genética , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Sumoilación/fisiología , Sustitución de Aminoácidos , Células HEK293 , Infecciones por VIH/virología , Humanos , Lisina , Mutación , Fosforilación , Proteína 1 que Contiene Dominios SAM y HD/química , Células U937
18.
Oncogene ; 40(12): 2243-2257, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33649538

RESUMEN

Mdm2 antagonizes the tumor suppressor p53. Targeting the Mdm2-p53 interaction represents an attractive approach for the treatment of cancers with functional p53. Investigating mechanisms underlying Mdm2-p53 regulation is therefore important. The scaffold protein ß-arrestin2 (ß-arr2) regulates tumor suppressor p53 by counteracting Mdm2. ß-arr2 nucleocytoplasmic shuttling displaces Mdm2 from the nucleus to the cytoplasm resulting in enhanced p53 signaling. ß-arr2 is constitutively exported from the nucleus, via a nuclear export signal, but mechanisms regulating its nuclear entry are not completely elucidated. ß-arr2 can be SUMOylated, but no information is available on how SUMO may regulate ß-arr2 nucleocytoplasmic shuttling. While we found ß-arr2 SUMOylation to be dispensable for nuclear import, we identified a non-covalent interaction between SUMO and ß-arr2, via a SUMO interaction motif (SIM), that is required for ß-arr2 cytonuclear trafficking. This SIM promotes association of ß-arr2 with the multimolecular RanBP2/RanGAP1-SUMO nucleocytoplasmic transport hub that resides on the cytoplasmic filaments of the nuclear pore complex. Depletion of RanBP2/RanGAP1-SUMO levels result in defective ß-arr2 nuclear entry. Mutation of the SIM inhibits ß-arr2 nuclear import, its ability to delocalize Mdm2 from the nucleus to the cytoplasm and enhanced p53 signaling in lung and breast tumor cell lines. Thus, a ß-arr2 SIM nuclear entry checkpoint, coupled with active ß-arr2 nuclear export, regulates its cytonuclear trafficking function to control the Mdm2-p53 signaling axis.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína SUMO-1/genética , Proteína p53 Supresora de Tumor/genética , Arrestina beta 2/genética , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Mutación/genética , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Señales de Exportación Nuclear/genética , Transducción de Señal/genética , Sumoilación/genética
19.
Virologie (Montrouge) ; 14(5): 349-361, 2010 Oct 01.
Artículo en Francés | MEDLINE | ID: mdl-36151696

RESUMEN

Post-translational modifications, such as SUMOylation, are exam- ples of cellular machineries hijacked by viruses to efficiently replicate. SUMOylation, which consists in the conjugation of small ubiquitin-like modi- fier (SUMO) peptides to a substrate, is exploited or hampered by numerous viruses during infection. Several viral proteins are SUMOylated, causing modulation of sub-cellular localization, stability or modifications of protein activities. In this review, recently described viral examples have been chosen to highlight the different strategies used by viruses to hijack SUMOylation in order to promote replication. The link between pathologies due to viral infec- tions and SUMOylation is discussed. Finally, the potential applications of SUMOylation inhibitors in the treatment of viral infections and associated cancer are evoked.

20.
Retrovirology ; 6: 26, 2009 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-19272132

RESUMEN

The rate of HIV-1 gene expression is a key step that determines the kinetics of virus spread and AIDS progression. Viral entry and gene expression were described to be the key determinants for cell permissiveness to HIV. Recent reports highlighted the involvement of miRNA in regulating HIV-1 replication post-transcriptionally. In this study we explored the role of cellular factors required for miRNA-mediated mRNA translational inhibition in regulating HIV-1 gene expression. Here we show that HIV-1 mRNAs associate and co-localize with components of the RNA Induced Silencing Complex (RISC), and we characterize some of the proteins required for miRNA-mediated silencing (miRNA effectors). RCK/p54, GW182, LSm-1 and XRN1 negatively regulate HIV-1 gene expression by preventing viral mRNA association with polysomes. Interestingly, knockdown of RCK/p54 or DGCR8 resulted in virus reactivation in PBMCs isolated from HIV infected patients treated with suppressive HAART.


Asunto(s)
Regulación Viral de la Expresión Génica , VIH-1/fisiología , MicroARNs/metabolismo , ARN Viral/metabolismo , Replicación Viral/fisiología , Proteínas Argonautas , Línea Celular , Células Cultivadas , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Polirribosomas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Latencia del Virus/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA