Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 487
Filtrar
1.
PLoS Pathog ; 18(2): e1009986, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35139135

RESUMEN

The Nrf2/Keap1 axis plays a complex role in viral susceptibility, virus-associated inflammation and immune regulation in host cells. However, whether or how the Nrf2/Keap1 axis is involved in the interactions between equine lentiviruses and their hosts remains unclear. Here, we demonstrate that the Nrf2/Keap1 axis was activated during EIAV infection. Mechanistically, EIAV-Rev competitively binds to Keap1 and releases Nrf2 from Keap1-mediated repression, leading to the accumulation of Nrf2 in the nucleus and promoting Nrf2 responsive genes transcription. Subsequently, we demonstrated that the Nrf2/Keap1 axis represses EIAV replication via two independent molecular mechanisms: directly increasing antioxidant enzymes to promote effective cellular resistance against EIAV infection, and repression of Rev-mediated RNA transport through direct interaction between Keap1 and Rev. Together, these data suggest that activation of the Nrf2/Keap1 axis mediates a passive defensive response to combat EIAV infection. The Nrf2/Keap1 axis could be a potential target for developing strategies for combating EIAV infection.


Asunto(s)
Antivirales/farmacología , Productos del Gen rev/metabolismo , Virus de la Anemia Infecciosa Equina/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Antioxidantes/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
2.
J Biol Chem ; 295(32): 11174-11183, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32554809

RESUMEN

Stimulator of interferon genes (STING) mediates cytosolic DNA-induced innate immune signaling via membrane trafficking. The global identification of proteins that spatiotemporally interact with STING will provide a better understanding of its trafficking mechanisms and of STING signaling pathways. Proximity-dependent biotin identification (BioID) is a powerful technology to identify physiologically relevant protein-protein interactions in living cells. However, biotinylated peptides are rarely detected in the conventional BioID method, which uses streptavidin beads to pull down biotinylated proteins, because the biotin-streptavidin interaction is too strong. As a result, only nonbiotinylated peptides are identified, which cannot be distinguished from peptides of nonspecifically pull-downed proteins. Here, we developed a simple method to efficiently and specifically enrich biotinylated peptides using Tamavidin 2-REV, an engineered avidin-like protein with reversible biotin-binding capability. Using RAW264.7 macrophages stably expressing TurboID-fused STING, we identified and quantified >4,000 biotinylated peptides of STING-proximal proteins. Various endoplasmic reticulum-associated proteins were biotinylated in unstimulated cells, and STING activation caused biotinylation of many proteins located in the Golgi and endosomes. These proteins included those known to interact with activated STING, such as TANK-binding kinase 1 (TBK1), several palmitoyl transferases, and p62/sequestosome 1 (SQSTM1). Furthermore, interferon-induced transmembrane protein 3 (IFITM3), an endolysosome-localized antiviral protein, bound to STING at the late activation stage. These dynamic interaction profiles will provide detailed insights into STING signaling; we propose that our approach using Tamavidin 2-REV would be useful for BioID-based and other biotinylation-based peptide identification methods.


Asunto(s)
Avidina/metabolismo , Productos del Gen rev/metabolismo , Proteínas de la Membrana/genética , Animales , Biotinilación , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Péptidos/metabolismo , Fosforilación , Células RAW 264.7 , Transducción de Señal
3.
Biomacromolecules ; 19(10): 3945-3957, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30160482

RESUMEN

The simultaneous expression in Escherichia coli cells of the Qß virus-like particle (VLP) capsid protein and protein "cargo" tagged with a positively charged Rev peptide sequence leads to the spontaneous self-assembly of VLPs with multiple copies of the cargo inside. We report the packaging of four new enzymes with potential applications in medicine and chemical manufacturing. The captured enzymes are active while inside the nanoparticle shell and are protected from environmental conditions that lead to free-enzyme destruction. We also describe genetic modifications to the packaging scheme that shed light on the self-assembly mechanism of this system and allow indirect control over the internal packaging density of cargo. The technology was extended to create, via self-assembly, VLPs that simultaneously display protein ligands on the exterior and contain enzymes within. Inverse relationships were observed between the size of both the packaged and externally displayed protein or domains and nanoparticle yield. These results provide a general method for the rapid creation of robust protein nanoparticles with desired catalytic and targeting functionalities.


Asunto(s)
Proteínas de la Cápside/metabolismo , Productos del Gen rev/metabolismo , Enzimas Multifuncionales/química , Enzimas Multifuncionales/metabolismo , Nanopartículas/metabolismo , ARN Viral/metabolismo , Ensamble de Virus , Aldehído-Liasas/química , Aldehído-Liasas/genética , Aldehído-Liasas/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Catálisis , Citosina Desaminasa/química , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Productos del Gen rev/química , Productos del Gen rev/genética , Células HeLa , Humanos , Enzimas Multifuncionales/genética , Nanopartículas/química , ARN Viral/química , ARN Viral/genética
4.
Retrovirology ; 14(1): 40, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28830558

RESUMEN

BACKGROUND: Rev-like proteins are post-transcriptional regulatory proteins found in several retrovirus genera, including lentiviruses, betaretroviruses, and deltaretroviruses. These essential proteins mediate the nuclear export of incompletely spliced viral RNA, and act by tethering viral pre-mRNA to the host CRM1 nuclear export machinery. Although all Rev-like proteins are functionally homologous, they share less than 30% sequence identity. In the present study, we computationally assessed the extent of structural homology among retroviral Rev-like proteins within a phylogenetic framework. RESULTS: We undertook a comprehensive analysis of overall protein domain architecture and predicted secondary structural features for representative members of the Rev-like family of proteins. Similar patterns of α-helical domains were identified for Rev-like proteins within each genus, with the exception of deltaretroviruses, which were devoid of α-helices. Coiled-coil oligomerization motifs were also identified for most Rev-like proteins, with the notable exceptions of HIV-1, the deltaretroviruses, and some small ruminant lentiviruses. In Rev proteins of primate lentiviruses, the presence of predicted coiled-coil motifs segregated within specific primate lineages: HIV-1 descended from SIVs that lacked predicted coiled-coils in Rev whereas HIV-2 descended from SIVs that contained predicted coiled-coils in Rev. Phylogenetic ancestral reconstruction of coiled-coils for all Rev-like proteins predicted a single origin for the coiled-coil motif, followed by three losses of the predicted signal. The absence of a coiled-coil signal in HIV-1 was associated with replacement of canonical polar residues with non-canonical hydrophobic residues. However, hydrophobic residues were retained in the key 'a' and 'd' positions, and the α-helical region of HIV-1 Rev oligomerization domain could be modeled as a helical wheel with two predicted interaction interfaces. Moreover, the predicted interfaces mapped to the dimerization and oligomerization interfaces in HIV-1 Rev crystal structures. Helical wheel projections of other retroviral Rev-like proteins, including endogenous sequences, revealed similar interaction interfaces that could mediate oligomerization. CONCLUSIONS: Sequence-based computational analyses of Rev-like proteins, together with helical wheel projections of oligomerization domains, reveal a conserved homogeneous structural basis for oligomerization by retroviral Rev-like proteins.


Asunto(s)
Productos del Gen rev/química , Productos del Gen rev/metabolismo , Modelos Moleculares , Retroviridae/química , Retroviridae/metabolismo , Secuencia de Aminoácidos , Dimerización , Variación Genética , Filogenia , Estructura Secundaria de Proteína , Proteínas de los Retroviridae/química , Proteínas de los Retroviridae/metabolismo , Homología de Secuencia de Aminoácido
5.
Proc Natl Acad Sci U S A ; 110(12): 4750-5, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23471982

RESUMEN

The mammalian circadian clock is composed of interlocking feedback loops. Cryptochrome is a central component in the core negative feedback loop, whereas Rev-Erbα, a member of the nuclear receptor family, is an essential component of the interlocking loop. To understand the roles of different clock genes, we conducted a genetic interaction screen by generating single- and double-mutant mice. We found that the deletion of Rev-erbα in F-box/leucine rich-repeat protein (Fbxl3)-deficient mice rescued its long-circadian period phenotype, and our results further revealed that FBXL3 regulates Rev-Erb/retinoic acid receptor-related orphan receptor-binding element (RRE)-mediated transcription by inactivating the Rev-Erbα:histone deacetylase 3 corepressor complex. By analyzing the Fbxl3 and Cryptochrome 1 double-mutant mice, we found that FBXL3 also regulates the amplitudes of E-box-driven gene expression. These two separate roles of FBXL3 in circadian feedback loops provide a mechanism that contributes to the period determination and robustness of the clock.


Asunto(s)
Ritmo Circadiano/fisiología , Criptocromos/metabolismo , Proteínas F-Box/metabolismo , Regulación de la Expresión Génica/fisiología , Animales , Criptocromos/genética , Proteínas F-Box/genética , Productos del Gen rev/genética , Productos del Gen rev/metabolismo , Ratones , Ratones Noqueados , Elementos de Respuesta/fisiología , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo
6.
J Virol ; 88(8): 4069-82, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478440

RESUMEN

UNLABELLED: Intron-containing mRNAs are subject to restricted nuclear export in higher eukaryotes. Retroviral replication requires the nucleocytoplasmic transport of both spliced and unspliced RNA transcripts, and RNA export mechanisms of gammaretroviruses are poorly characterized. Here, we report the involvement of the nuclear export receptor NXF1/TAP in the nuclear export of gammaretroviral RNA transcripts. We identified a conserved cis-acting element in the pol gene of gammaretroviruses, including murine leukemia virus (MLV) and xenotropic murine leukemia virus (XMRV), named the CAE (cytoplasmic accumulation element). The CAE enhanced the cytoplasmic accumulation of viral RNA transcripts and the expression of viral proteins without significantly affecting the stability, splicing, or translation efficiency of the transcripts. Insertion of the CAE sequence also facilitated Rev-independent HIV Gag expression. We found that the CAE sequence interacted with NXF1, whereas disruption of NXF1 ablated CAE function. Thus, the CAE sequence mediates the cytoplasmic accumulation of gammaretroviral transcripts in an NXF1-dependent manner. Disruption of NXF1 expression impaired cytoplasmic accumulations of both spliced and unspliced RNA transcripts of XMRV and MLV, resulting in their nuclear retention or degradation. Thus, our results demonstrate that gammaretroviruses use NXF1 for the cytoplasmic accumulation of both spliced and nonspliced viral RNA transcripts. IMPORTANCE: Murine leukemia virus (MLV) has been studied as one of the classic models of retrovirology. Although unspliced host messenger RNAs are rarely exported from the nucleus, MLV actively exports unspliced viral RNAs to the cytoplasm. Despite extensive studies, how MLV achieves this difficult task has remained a mystery. Here, we have studied the RNA export mechanism of MLV and found that (i) the genome contains a sequence which supports the efficient nuclear export of viral RNAs, (ii) the cellular factor NXF1 is involved in the nuclear export of both spliced and unspliced viral RNAs, and, finally, (iii) depletion of NXF1 results in nuclear retention or degradation of viral RNAs. Our study provides a novel insight into MLV nuclear export.


Asunto(s)
Virus de la Leucemia Murina/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Empalme del ARN , ARN Viral/metabolismo , Infecciones por Retroviridae/veterinaria , Enfermedades de los Roedores/metabolismo , Transporte Activo de Núcleo Celular , Animales , Secuencia de Bases , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/virología , Productos del Gen rev/genética , Productos del Gen rev/metabolismo , Virus de la Leucemia Murina/genética , Ratones , Datos de Secuencia Molecular , Proteínas de Transporte Nucleocitoplasmático/genética , ARN Viral/genética , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/virología
7.
Org Biomol Chem ; 13(6): 1792-9, 2015 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-25503645

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) Rev protein facilitates the export of viral RNA from nucleus to cytoplasm, which is a key step in HIV-1 pathogenesis and transmission. In this study, we have screened a commercial library and identified the hit compound 1 bearing a benzenesulfonamide quinoline scaffold that inhibited Rev activity and HIV-1 infectivity. Compounds bearing this scaffold were synthesized and their SAR was studied. We identified compound 20 with low toxicity and potent activity to inhibit HIV-1 replication by affecting Rev function.


Asunto(s)
Fármacos Anti-VIH/farmacología , Derivados del Benceno/farmacología , Productos del Gen rev/antagonistas & inhibidores , VIH-1/efectos de los fármacos , Quinolinas/farmacología , Sulfonamidas/farmacología , Replicación Viral/efectos de los fármacos , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/química , Derivados del Benceno/síntesis química , Derivados del Benceno/química , Productos del Gen rev/metabolismo , VIH-1/crecimiento & desarrollo , VIH-1/metabolismo , Estructura Molecular , Quinolinas/síntesis química , Quinolinas/química , Sulfonamidas/síntesis química , Sulfonamidas/química
8.
Retrovirology ; 11: 115, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25533001

RESUMEN

BACKGROUND: The lentiviral Rev protein mediates nuclear export of intron-containing viral RNAs that encode structural proteins or serve as the viral genome. Following translation, HIV-1 Rev localizes to the nucleus and binds its cognate sequence, termed the Rev-responsive element (RRE), in incompletely spliced viral RNA. Rev subsequently multimerizes along the viral RNA and associates with the cellular Crm1 export machinery to translocate the RNA-protein complex to the cytoplasm. Equine infectious anemia virus (EIAV) Rev is functionally homologous to HIV-1 Rev, but shares very little sequence similarity and differs in domain organization. EIAV Rev also contains a bipartite RNA binding domain comprising two short arginine-rich motifs (designated ARM-1 and ARM-2) spaced 79 residues apart in the amino acid sequence. To gain insight into the topology of the bipartite RNA binding domain, a computational approach was used to model the tertiary structure of EIAV Rev. RESULTS: The tertiary structure of EIAV Rev was modeled using several protein structure prediction and model quality assessment servers. Two types of structures were predicted: an elongated structure with an extended central alpha helix, and a globular structure with a central bundle of helices. Assessment of models on the basis of biophysical properties indicated they were of average quality. In almost all models, ARM-1 and ARM-2 were spatially separated by >15 Å, suggesting that they do not form a single RNA binding interface on the monomer. A highly conserved canonical coiled-coil motif was identified in the central region of EIAV Rev, suggesting that an RNA binding interface could be formed through dimerization of Rev and juxtaposition of ARM-1 and ARM-2. In support of this, purified Rev protein migrated as a dimer in Blue native gels, and mutation of a residue predicted to form a key coiled-coil contact disrupted dimerization and abrogated RNA binding. In contrast, mutation of residues outside the predicted coiled-coil interface had no effect on dimerization or RNA binding. CONCLUSIONS: Our results suggest that EIAV Rev binding to the RRE requires dimerization via a coiled-coil motif to juxtapose two RNA binding motifs, ARM-1 and ARM-2.


Asunto(s)
Productos del Gen rev/química , Productos del Gen rev/metabolismo , Virus de la Anemia Infecciosa Equina/fisiología , Multimerización de Proteína , ARN Viral/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica
9.
J Virol ; 86(9): 4892-905, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22379104

RESUMEN

The Rev protein is essential for the replication of lentiviruses. Rev is a shuttling protein that transports unspliced and partially spliced lentiviral RNAs from the nucleus to the cytoplasm via the nucleopore. To transport these RNAs, the human immunodeficiency virus type 1 (HIV-1) Rev uses the karyopherin ß family importin ß and CRM1 proteins that interact with the Rev nuclear localization signal (NLS) and nuclear exportation signal (NES), respectively. Recently, we reported the presence of new types of bipartite NLS and nucleolar localization signal (NoLS) in the bovine immunodeficiency virus (BIV) Rev protein. Here we report the characterization of the nuclear import and export pathways of BIV Rev. By using an in vitro nuclear import assay, we showed that BIV Rev is transported into the nucleus by a cytosolic and energy-dependent importin α/ß classical pathway. Results from glutathione S-transferase (GST) pulldown assays that showed the binding of BIV Rev with importins α3 and α5 were in agreement with those from the nuclear import assay. We also identified a leptomycin B-sensitive NES in BIV Rev, which indicates that the protein is exported via CRM1 like HIV-1 Rev. Mutagenesis experiments showed that the BIV Rev NES maps between amino acids 109 to 121 of the protein. Remarkably, the BIV Rev NES was found to be of the cyclic AMP (cAMP)-dependent protein kinase inhibitor (PKI) type instead of the HIV-1 Rev type. In summary, our data showed that the nuclear import mechanism of BIV Rev is novel among Rev proteins characterized so far in lentiviruses.


Asunto(s)
Productos del Gen rev/química , Productos del Gen rev/metabolismo , Virus de la Inmunodeficiencia Bovina/metabolismo , Señales de Exportación Nuclear , Transporte Activo de Núcleo Celular/fisiología , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Bovinos , Ácidos Grasos Insaturados/farmacología , Humanos , Carioferinas/antagonistas & inhibidores , Poro Nuclear/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Transducción de Señal , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Proteína de Unión al GTP ran/metabolismo , Proteína Exportina 1
10.
Proc Natl Acad Sci U S A ; 107(28): 12481-6, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20616058

RESUMEN

RNA is a crucial structural component of many ribonucleoprotein (RNP) complexes, including the ribosome, spliceosome, and signal recognition particle, but the role of RNA in guiding complex formation is only beginning to be explored. In the case of HIV, viral replication requires assembly of an RNP composed of the Rev protein homooligomer and the Rev response element (RRE) RNA to mediate nuclear export of unspliced viral mRNAs. Assembly of the functional Rev-RRE complex proceeds by cooperative oligomerization of Rev on the RRE scaffold and utilizes both protein-protein and protein-RNA interactions to organize complexes with high specificity. The structures of the Rev protein and a peptide-RNA complex are known, but the complete RNP is not, making it unclear to what extent RNA defines the composition and architecture of Rev-RNA complexes. Here we show that the RRE controls the oligomeric state and solubility of Rev and guides its assembly into discrete Rev-RNA complexes. SAXS and EM data were used to derive a structural model of a Rev dimer bound to an essential RRE hairpin and to visualize the complete Rev-RRE RNP, demonstrating that RRE binding drives assembly of Rev homooligomers into asymmetric particles, reminiscent of the role of RNA in organizing more complex RNP machines, such as the ribosome, composed of many different protein subunits. Thus, the RRE is not simply a passive scaffold onto which proteins bind but instead actively defines the protein composition and organization of the RNP.


Asunto(s)
Productos del Gen rev/química , Productos del Gen rev/metabolismo , VIH/genética , Citoplasma/genética , Citoplasma/metabolismo , Productos del Gen rev/genética , VIH/metabolismo , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Péptidos/genética , Péptidos/metabolismo , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elementos de Respuesta , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Dispersión del Ángulo Pequeño , Replicación Viral/genética
11.
PLoS Comput Biol ; 7(12): e1002309, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22194677

RESUMEN

The circadian clock is accountable for the regulation of internal rhythms in most living organisms. It allows the anticipation of environmental changes during the day and a better adaptation of physiological processes. In mammals the main clock is located in the suprachiasmatic nucleus (SCN) and synchronizes secondary clocks throughout the body. Its molecular constituents form an intracellular network which dictates circadian time and regulates clock-controlled genes. These clock-controlled genes are involved in crucial biological processes including metabolism and cell cycle regulation. Its malfunction can lead to disruption of biological rhythms and cause severe damage to the organism. The detailed mechanisms that govern the circadian system are not yet completely understood. Mathematical models can be of great help to exploit the mechanism of the circadian circuitry. We built a mathematical model for the core clock system using available data on phases and amplitudes of clock components obtained from an extensive literature search. This model was used to answer complex questions for example: how does the degradation rate of Per affect the period of the system and what is the role of the ROR/Bmal/REV-ERB (RBR) loop? Our findings indicate that an increase in the RNA degradation rate of the clock gene Period (Per) can contribute to increase or decrease of the period--a consequence of a non-monotonic effect of Per transcript stability on the circadian period identified by our model. Furthermore, we provide theoretical evidence for a potential role of the RBR loop as an independent oscillator. We carried out overexpression experiments on members of the RBR loop which lead to loss of oscillations consistent with our predictions. These findings challenge the role of the RBR loop as a merely auxiliary loop and might change our view of the clock molecular circuitry and of the function of the nuclear receptors (REV-ERB and ROR) as a putative driving force of molecular oscillations.


Asunto(s)
Relojes Circadianos , Modelos Biológicos , Proteínas Circadianas Period/metabolismo , Animales , Proteínas CLOCK/metabolismo , Productos del Gen rev/metabolismo , Humanos , Mamíferos , Ratones , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Núcleo Supraquiasmático/metabolismo
12.
Sci Rep ; 12(1): 18416, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36319640

RESUMEN

During HIV infection, intron-containing viral mRNAs are exported from the cell nucleus to the cytoplasm to complete the replication cycle. Cellular restrictions on the export of incompletely spliced transcripts are overcome by a viral protein, Rev, and an RNA structure found in all unspliced and incompletely spliced viral mRNAs, the Rev Response Element (RRE). Primary HIV isolates display substantial variation in the sequence and functional activity of Rev proteins. We analyzed Rev from two primary isolates with disparate activity that resulted in differences in in vitro fitness of replication-competent viral constructs. The results showed that amino acid differences within the oligomerization domain, but not the arginine-rich motif or the nuclear export signal, determined the level of Rev activity. Two specific amino acid substitutions were sufficient to alter the low-activity Rev to a high-activity phenotype. Other mutations in Rev sequences had unpredictable effects on activity that differed between the two Rev backbones. The sensitivity of Rev function level to small sequence changes likely permits modulation of Rev-RRE activity during HIV infection, which may play a role in pathogenesis. The functional consequences of Rev mutations differed between primary isolates, highlighting the challenge of generalizing studies of Rev conducted using laboratory HIV strains.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , VIH-1/genética , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , Infecciones por VIH/genética , Productos del Gen rev/genética , Productos del Gen rev/metabolismo , Elementos de Respuesta , Seropositividad para VIH/genética , ARN Mensajero/genética , ARN Viral/genética
13.
Virology ; 576: 30-41, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36137490

RESUMEN

Rev is an essential regulatory protein of Human Immunodeficiency Virus type 1 (HIV) that is found in the nucleus of infected cells. Rev multimerizes on the Rev-response element (RRE) of HIV RNA to facilitate the export of intron-containing HIV mRNAs from the nucleus to the cytoplasm, and, as such, HIV cannot replicate in the absence of Rev. We have developed cell-intact and cell-free assays based upon a robust firefly split-luciferase complementation system, both of which quantify Rev-Rev interaction. Using the cell-based system we show that additional Crm1 did not impact the interaction, whereas excess Rev reduced it. Furthermore, when a series of mutant Revs were tested, there was a strong correlation between the results of the cell-based assay and the results of a functional Rev trans-complementation infectivity assay. Of interest, a camelid nanobody (NB) that was known to inhibit Rev function enhanced Rev-Rev interaction in the cell-based system. We observed a similar increase in Rev-Rev interaction in a cell-free system, when cell lysates expressing Rev-NLUC or CLUC-Rev were simply mixed. In the cell-free system Rev-Rev interaction occurred within minutes and was inhibited by excess Rev. The levels of interaction between the mutant Revs tested varied by mutant type. Treatment of Rev lysates with RNAse minimally reduced the degree of interaction whereas addition of HIV RRE RNA enhanced the interaction. Purified GST-Rev protein inhibited the interaction. The Z-factor (Z') for the cell-free system was ∼0.85 when tested in 96-well format, and the anti-Rev NB enhanced the interaction in the cell-free system. Thus, we have developed both cell-intact and cell-free systems that can reliably, rapidly, and reproducibly quantify Rev-Rev interaction. These assays, particularly the cell-free one, may be useful in screening and identifying compounds that inhibit Rev function on a high throughput basis.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/metabolismo , VIH-1/fisiología , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Productos del Gen rev/genética , Productos del Gen rev/metabolismo , ARN/metabolismo , Ribonucleasas/metabolismo , ARN Viral/genética
14.
J Biol Chem ; 285(28): 21768-80, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20406803

RESUMEN

The human immunodeficiency virus, type 1 (HIV-1)-encoded Rev protein is essential for the expression of late viral mRNAs. Rev forms a large organized multimeric protein-protein complex on the Rev response element of these viral mRNA species and transports them from the nucleus to the cytoplasm, exploiting the CRM1-mediated cellular machinery. Here we report the selection of a nanobody, derived from a llama heavy-chain only antibody, that efficiently blocks the assembly of Rev multimers. The nanobody inhibits HIV-1 replication in cells and specifically suppresses the Rev-dependent expression of partially spliced and unspliced HIV-1 RNA. In HIV-susceptible cells, this nanobody thus has potential as an effective anti-HIV agent using genetic immunization strategies. Its binding site was mapped to Rev residues Lys-20 and Tyr-23 located in the N-terminal alpha-helical multimerization domain. In the presence of this nanobody, we observed an accumulation of dimeric Rev species, supporting a head-to-head/tail-to-tail molecular model for Rev assembly. The results indicate that the oligomeric assembly of Rev follows an ordered stepwise process and identify a new epitope within Rev that could guide strategies for the development of novel HIV inhibitors.


Asunto(s)
Productos del Gen rev/química , Infecciones por VIH/tratamiento farmacológico , VIH-1/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Fármacos Anti-VIH/farmacología , Anticuerpos/química , Camélidos del Nuevo Mundo , Dimerización , Transferencia Resonante de Energía de Fluorescencia , Productos del Gen rev/metabolismo , Células HeLa , Humanos , Lisina/química , Datos de Secuencia Molecular , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Tirosina/química
16.
RNA Biol ; 8(2): 316-24, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21358282

RESUMEN

The export of viral RNA from the nucleus to the cytoplasm of the cellular host is a crucial step in the life cycle of HIV-1 that is mediated by the viral Rev protein. One aspect of the Rev function, its multimerization, is still unexplored as a target for antiviral therapy. This is partly due to the lack of a fast and solid system to measure Rev multimerization. We have developed a high throughput in vitro Rev multimerization assay based on fluorescence resonance energy transfer (FRET) in which real-time Rev-Rev interactions can be measured both in the absence and the presence of Rev specific RRE RNA. Well-characterized Rev multimerization deficient mutants showed reduced FRET as well as unlabeled Rev molecules were able to inhibit the FRET signal demonstrating the specificity of the assay. Upon multimerization along RRE RNA the FRET signal significantly increased but dropped again at equimolar Rev/RRE ratios suggesting that in this condition most Rev molecules are bound as monomers to the RRE. Furthermore, using this assay, we demonstrate that a previously selected llama heavy-chain only antibody was shown to not only prevent the development of Rev multimers but also disassemble the already formed complexes confirming the dynamic nature of the Rev-Rev interactions. The in vitro FRET based multimerization assay facilitates the further study of the basic mechanism of cooperative Rev multimerization along the RRE and is also widely applicable to study the assembly of other functional complexes involving protein homo-multimerization or cooperative protein-protein interactions on RNA or DNA.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Productos del Gen rev/metabolismo , VIH-1/metabolismo , Multimerización de Proteína , Productos del Gen rev/genética , Genes env/genética , VIH-1/genética , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Transporte de ARN , ARN Viral/genética
17.
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
18.
RNA Biol ; 8(2): 325-42, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21368586

RESUMEN

HIV-1 pre-mRNA splicing depends upon 4 donor and 8 acceptor sites, which are used in combination to produce more than 40 different mRNAs. The acceptor site A7 plays an essential role for tat and rev mRNA production. The SLS2-A7 stem-loop structure containing site A7 was also proposed to modulate HIV-1 RNA export by the Rev protein. To further characterize nuclear factors involved in these processes, we purified RNP complexes formed by incubation of SLS2-A7 RNA transcripts in HeLa cell nuclear extracts by affinity chromatography and identified 33 associated proteins by nanoLC-MS/MS. By UV cross-linking, immunoselection and EMSA, we showed that, in addition to the well-known hnRNP A1 inhibitor of site A7, nucleolin, hnRNP H and hnRNP K interact directly with SLS2-A7 RNA. Nucleolin binds to a cluster of successive canonical NRE motifs in SLS2-A7 RNA, which is unique in HIV-1 RNA. Proteins hnRNP A1 and hnRNP K bind synergistically to SLS2-A7 RNA and both have a negative effect on site A7 activity. By the use of a plasmid expressing a truncated version of HIV-1 RNA, we showed a strong effect of the overexpression of hnRNP K in HeLa cells on HIV-1 alternative splicing. As a consequence, production of the Nef protein was strongly reduced. Interestingly also, many proteins identified in our proteomic analysis are known to modulate either the Rev activity or other mechanisms required for HIV-1 multiplication and several of them seem to be recruited by hnRNP K, suggesting that hnRNP K plays an important role for HIV-1 biology.


Asunto(s)
Productos del Gen rev/metabolismo , VIH-1/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Empalme del ARN , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Empalme Alternativo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Exones , Regulación Viral de la Expresión Génica , Productos del Gen rev/genética , VIH-1/metabolismo , Células HeLa , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Fosfoproteínas/metabolismo , Unión Proteica , Precursores del ARN/genética , ARN Viral/genética , ARN Viral/metabolismo , ARN Viral/ultraestructura , Proteínas de Unión al ARN/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/biosíntesis , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Nucleolina
19.
J Exp Med ; 176(4): 1197-201, 1992 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-1402661

RESUMEN

The human immunodeficiency virus (HIV) Rev protein is essential for viral structural protein expression (Gag, Pol, and Env) and, hence, for viral replication. In transient transfection assays, mutant forms of Rev have been identified that inhibit wild-type Rev activity and therefore suppress viral replication. To determine whether such transdominant Rev proteins could provide long-term protection against HIV infection without affecting T cell function, T leukemia cell lines were stably transduced with a retroviral vector encoding a transdominant mutant of the Rev protein, M10. While all the M10-expressing cell lines remained infectable by HIV-1, these same cells failed to support a productive replication cycle when infected with a cloned isolate of HIV-1. In addition, two out of three M10-expressing CEM clones were also resistant to highly productive infection by a heterogeneous HIV-1 pool. Expression of M10 did not affect induction of HIV transcription mediated by the kappa B regulatory element or Tat. Importantly, constitutive expression of Rev M10 did not alter the secretion of interleukin 2 in response to mitogen stimulation of EL-4 and Jurkat cells. The inhibition of HIV infection in cells stably expressing a transdominant Rev protein, in the absence of any deleterious effect on T cell function, suggests that such a strategy could provide a therapeutic effect in the T lymphocytes of acquired immunodeficiency syndrome patients.


Asunto(s)
Productos del Gen rev/metabolismo , VIH-1/fisiología , Linfocitos T/fisiología , Replicación Viral , Células Clonales , Productos del Gen rev/genética , VIH-1/genética , Humanos , Interleucina-2/biosíntesis , Activación de Linfocitos , FN-kappa B/metabolismo , Linfocitos T/microbiología , Transcripción Genética , Transfección , Productos del Gen rev del Virus de la Inmunodeficiencia Humana
20.
J Virol ; 83(19): 10280-5, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19605480

RESUMEN

Understanding the correlates of immune protection against human immunodeficiency virus and simian immunodeficiency virus (SIV) will require defining the entire cellular immune response against the viruses. Here, we define two novel translation products from the SIV env mRNA that are targeted by the T-cell response in SIV-infected rhesus macaques. The shorter product is a subset of the larger product, which contains both the first exon of the Rev protein and a translated portion of the rev intron. Our data suggest that the translation of viral alternate reading frames may be an important source of T-cell epitopes, including epitopes normally derived from functional proteins.


Asunto(s)
Epítopos de Linfocito T/química , Regulación Viral de la Expresión Génica , Productos del Gen rev/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Secuencia de Aminoácidos , Animales , Epítopos/química , Genes env , Macaca , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA