Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 39(2): 110690, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417684

RESUMEN

Viruses must effectively remodel host cellular pathways to replicate and evade immune defenses, and they must do so with limited genomic coding capacity. Targeting post-translational modification (PTM) pathways provides a mechanism by which viruses can broadly and rapidly transform a hostile host environment into a hospitable one. We use mass spectrometry-based proteomics to quantify changes in protein abundance and two PTM types-phosphorylation and ubiquitination-in response to HIV-1 infection with viruses harboring targeted deletions of a subset of HIV-1 genes. PTM analysis reveals a requirement for Aurora kinase activity in HIV-1 infection and identified putative substrates of a phosphatase that is degraded during infection. Finally, we demonstrate that the HIV-1 Vpr protein inhibits histone H1 ubiquitination, leading to defects in DNA repair.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH-1 , VIH-1/genética , Humanos , Procesamiento Proteico-Postraduccional , Proteómica , Ubiquitinación
2.
Mol Cell ; 78(2): 197-209.e7, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32084337

RESUMEN

We have developed a platform for quantitative genetic interaction mapping using viral infectivity as a functional readout and constructed a viral host-dependency epistasis map (vE-MAP) of 356 human genes linked to HIV function, comprising >63,000 pairwise genetic perturbations. The vE-MAP provides an expansive view of the genetic dependencies underlying HIV infection and can be used to identify drug targets and study viral mutations. We found that the RNA deadenylase complex, CNOT, is a central player in the vE-MAP and show that knockout of CNOT1, 10, and 11 suppressed HIV infection in primary T cells by upregulating innate immunity pathways. This phenotype was rescued by deletion of IRF7, a transcription factor regulating interferon-stimulated genes, revealing a previously unrecognized host signaling pathway involved in HIV infection. The vE-MAP represents a generic platform that can be used to study the global effects of how different pathogens hijack and rewire the host during infection.


Asunto(s)
Epistasis Genética , Infecciones por VIH/genética , Factor 7 Regulador del Interferón/genética , Factores de Transcripción/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Interferones/genética , Mutación , Transducción de Señal/genética
3.
Cell Host Microbe ; 26(1): 86-99.e7, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31253590

RESUMEN

The Cullin-RING E3 ligase (CRL) family is commonly hijacked by pathogens to redirect the host ubiquitin proteasome machinery to specific targets. During HIV infection, CRL5 is hijacked by HIV Vif to target viral restriction factors of the APOBEC3 family for ubiquitination and degradation. Here, using a quantitative proteomics approach, we identify the E3 ligase ARIH2 as a regulator of CRL5-mediated APOBEC3 degradation. The CUL5Vif/CBFß complex recruits ARIH2 where it acts to transfer ubiquitin directly to the APOBEC3 targets. ARIH2 is essential for CRL5-dependent HIV infectivity in primary CD4+ T cells. Furthermore, we show that ARIH2 cooperates with CRL5 to prime other cellular substrates for polyubiquitination, suggesting this may represent a general mechanism beyond HIV infection and APOBEC3 degradation. Taken together, these data identify ARIH2 as a co-factor in the Vif-hijacked CRL5 complex that contributes to HIV infectivity and demonstrate the operation of the E1-E2-E3/E3-substrate ubiquitination mechanism in a viral infection context.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , Proteínas Cullin/metabolismo , Infecciones por VIH/patología , Interacciones Huésped-Patógeno , Evasión Inmune , Ubiquitina-Proteína Ligasas/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Humanos , Modelos Teóricos , Proteolisis , Proteoma/análisis , Replicación Viral
4.
Artículo en Inglés | MEDLINE | ID: mdl-28416550

RESUMEN

Viral regulatory complexes perform critical functions during virus replication and are important targets for therapeutic intervention. In HIV, the Tat and Rev proteins form complexes with multiple viral and cellular factors to direct transcription and export of the viral RNA. These complexes are composed of many proteins and are dynamic, making them difficult to fully recapitulate in vitro Therefore, we developed a cell-based reporter assay to monitor the assembly of viral complexes for inhibitor screening. We screened a small-molecule library and identified multiple hits that inhibit the activity of the viral complexes. A subsequent chemistry effort was focused on a thieno[2,3-b]pyridine scaffold, examples of which inhibited HIV replication and the emergence from viral latency. Notable aspects of the effort to determine the structure-activity relationship (SAR) include migration to the regioisomeric thieno[2,3-c]pyridine ring system and the identification of analogs with single-digit nanomolar activity in both reporter and HIV infectivity assays, an improvement of >100-fold in potency over the original hits. These results validate the screening strategy employed and reveal a promising lead series for the development of a new class of HIV therapeutics.


Asunto(s)
Fármacos Anti-VIH/farmacología , Antivirales/uso terapéutico , Piridinas/uso terapéutico , Regulación Viral de la Expresión Génica/genética , ARN Viral/genética , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
5.
Cell ; 167(7): 1762-1773.e12, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984726

RESUMEN

Overlapping genes pose an evolutionary dilemma as one DNA sequence evolves under the selection pressures of multiple proteins. Here, we perform systematic statistical and mutational analyses of the overlapping HIV-1 genes tat and rev and engineer exhaustive libraries of non-overlapped viruses to perform deep mutational scanning of each gene independently. We find a "segregated" organization in which overlapped sites encode functional residues of one gene or the other, but never both. Furthermore, this organization eliminates unfit genotypes, providing a fitness advantage to the population. Our comprehensive analysis reveals the extraordinary manner in which HIV minimizes the constraint of overlapping genes and repurposes that constraint to its own advantage. Thus, overlaps are not just consequences of evolutionary constraints, but rather can provide population fitness advantages.


Asunto(s)
Evolución Biológica , VIH-1/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Entropía , Aptitud Genética , Infecciones por VIH/virología , Humanos , Mutación , Sistemas de Lectura Abierta , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética
6.
Nat Prod Commun ; 10(1): 117-24, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25920233

RESUMEN

Human immunodeficiency virus (HIV) type I integrase (IN) active site, and viral DNA-binding residues K156 and K159 are predicted to interact both with strand transfer-selective IN inhibitors (STI), e.g. L-731,988, Elvitegravir (EVG), and the FDA-approved IN inhibitor, Raltegravir (RGV), and strand transfer non-selective inhibitors, e.g. dicaffeoyltartaric acids (DCTAs), e.g. L-chicoric acid (L-CA). To test posited roles for these two lysine residues in inhibitor action we assayed the potency of L-CA and several STI against a panel of K156 and K159 mutants. Mutagenesis of K156 conferred resistance to L-CA and mutagenesis of either K156 or K159 conferred resistance to STI indicating that the cationic charge at these two viral DNA-binding residues is important for inhibitor potency. IN K156N, a reported polymorphism associated with resistance to RGV, conferred resistance to L-CA and STI as well. To investigate the apparent preference L-CA exhibits for interactions with K156, we assayed the potency of several hybrid inhibitors containing combinations of DCTA and STI pharmacophores against recombinant IN K156A or K159A. Although K156A conferred resistance to diketo acid-branched bis-catechol hybrid inhibitors, neither K156A nor K159A conferred resistance to their monocatechol counterparts, suggesting that bis-catechol moieties direct DCTAs toward K156. In contrast, STI were more promiscuous in their interaction with K156 and K159. Taken together, the results of this study indicate that DCTAs interact with IN in a manner different than that of STI and suggest that DCTAs are an attractive candidate chemotype for development into drugs potent against STI-resistant IN.


Asunto(s)
Ácidos Cafeicos/farmacología , Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/efectos de los fármacos , Succinatos/farmacología , Integrasa de VIH/genética , Inhibidores de Integrasa VIH/química , Mutagénesis Sitio-Dirigida , Plantas Medicinales/química , Relación Estructura-Actividad
7.
Elife ; 3: e04120, 2014 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-25486594

RESUMEN

The HIV-1 protein Rev controls a critical step in viral replication by mediating the nuclear export of unspliced and singly-spliced viral mRNAs. Multiple Rev subunits assemble on the Rev Response Element (RRE), a structured region present in these RNAs, and direct their export through the Crm1 pathway. Rev-RRE assembly occurs via several Rev oligomerization and RNA-binding steps, but how these steps are coordinated to form an export-competent complex is unclear. Here, we report the first crystal structure of a Rev dimer-RRE complex, revealing a dramatic rearrangement of the Rev-dimer upon RRE binding through re-packing of its hydrophobic protein-protein interface. Rev-RNA recognition relies on sequence-specific contacts at the well-characterized IIB site and local RNA architecture at the second site. The structure supports a model in which the RRE utilizes the inherent plasticity of Rev subunit interfaces to guide the formation of a functional complex.


Asunto(s)
VIH-1/genética , ARN Viral/genética , Elementos de Respuesta , Replicación Viral/genética , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , Transporte Activo de Núcleo Celular , Sitios de Unión , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/virología , Cristalografía por Rayos X , Citosol/metabolismo , Citosol/virología , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Células HEK293 , VIH-1/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Modelos Moleculares , Unión Proteica , Empalme del ARN , ARN Viral/química , ARN Viral/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/virología , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/química , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/metabolismo , Proteína Exportina 1
8.
Mol Cell ; 49(4): 632-44, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23333304

RESUMEN

The HIV-1 accessory protein Vif hijacks a cellular Cullin-RING ubiquitin ligase, CRL5, to promote degradation of the APOBEC3 (A3) family of restriction factors. Recently, the cellular transcription cofactor CBFß was shown to form a complex with CRL5-Vif and to be essential for A3 degradation and viral infectivity. We now demonstrate that CBFß is required for assembling a well-ordered CRL5-Vif complex by inhibiting Vif oligomerization and by activating CRL5-Vif via direct interaction. The CRL5-Vif-CBFß holoenzyme forms a well-defined heterohexamer, indicating that Vif simultaneously hijacks CRL5 and CBFß. Heterodimers of CBFß and RUNX transcription factors contribute toward the regulation of genes, including those with immune system functions. We show that binding of Vif to CBFß is mutually exclusive with RUNX heterodimerization and impacts the expression of genes whose regulatory domains are associated with RUNX1. Our results provide a mechanism by which a pathogen with limited coding capacity uses one factor to hijack multiple host pathways.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Citosina Desaminasa/metabolismo , Regulación de la Expresión Génica , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasas APOBEC , Secuencia de Aminoácidos , Secuencia de Bases , Factor de Unión a CCAAT/química , Factor de Unión a CCAAT/fisiología , Secuencia de Consenso , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Citidina Desaminasa , Citosina Desaminasa/química , Citosina Desaminasa/fisiología , Expresión Génica , Genes Reporteros , Células HEK293 , VIH-1/fisiología , Interacciones Huésped-Patógeno , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Linfocitos T/metabolismo , Linfocitos T/virología , Ubiquitinación , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/fisiología
9.
PLoS Pathog ; 8(12): e1003085, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23300442

RESUMEN

Cellular restriction factors help to defend humans against human immunodeficiency virus (HIV). HIV accessory proteins hijack at least three different Cullin-RING ubiquitin ligases, which must be activated by the small ubiquitin-like protein NEDD8, in order to counteract host cellular restriction factors. We found that conjugation of NEDD8 to Cullin-5 by the NEDD8-conjugating enzyme UBE2F is required for HIV Vif-mediated degradation of the host restriction factor APOBEC3G (A3G). Pharmacological inhibition of the NEDD8 E1 by MLN4924 or knockdown of either UBE2F or its RING-protein binding partner RBX2 bypasses the effect of Vif, restoring the restriction of HIV by A3G. NMR mapping and mutational analyses define specificity determinants of the UBE2F NEDD8 cascade. These studies demonstrate that disrupting host NEDD8 cascades presents a novel antiretroviral therapeutic approach enhancing the ability of the immune system to combat HIV.


Asunto(s)
Proteínas Cullin/metabolismo , Citidina Desaminasa/metabolismo , VIH/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/antagonistas & inhibidores , Desaminasa APOBEC-3G , Linfocitos T CD4-Positivos/virología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Ciclopentanos/farmacología , Células HEK293 , VIH/crecimiento & desarrollo , Infecciones por VIH/inmunología , Humanos , Imagen por Resonancia Magnética , Proteína NEDD8 , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Ubiquitina-Proteína Ligasas/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
10.
J Med Chem ; 53(22): 8161-75, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20977258

RESUMEN

Fourteen analogues of the anti-HIV-1 integrase (IN) inhibitor L-chicoric acid (L-CA) were prepared. Their IC(50) values for 3'-end processing and strand transfer against recombinant HIV-1 IN were determined in vitro, and their cell toxicities and EC(50) against HIV-1 were measured in cells (ex vivo). Compounds 1-6 are catechol/ß-diketoacid hybrids, the majority of which exhibit submicromolar potency against 3'-end processing and strand transfer, though only with modest antiviral activities. Compounds 7-10 are L-CA/p-fluorobenzylpyrroloyl hybrids, several of which were more potent against strand transfer than 3'-end processing, a phenomenon previously attributed to the ß-diketo acid pharmacophore. Compounds 11-14 are tetrazole bioisosteres of L-CA and its analogues, whose in vitro potencies were comparable to L-CA but with enhanced antiviral potency. The trihydroxyphenyl analogue 14 was 30-fold more potent than L-CA at relatively nontoxic concentrations. These data indicate that L-CA analogues are attractive candidates for development into clinically relevant inhibitors of HIV-1 IN.


Asunto(s)
Ácidos Cafeicos/síntesis química , Inhibidores de Integrasa VIH/síntesis química , Integrasa de VIH/metabolismo , VIH-1/efectos de los fármacos , Cetoácidos/síntesis química , Succinatos/síntesis química , Tetrazoles/síntesis química , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacología , Línea Celular , Inhibidores de Integrasa VIH/química , Inhibidores de Integrasa VIH/farmacología , VIH-1/enzimología , Humanos , Cetoácidos/química , Cetoácidos/farmacología , Relación Estructura-Actividad , Succinatos/química , Succinatos/farmacología , Tetrazoles/química , Tetrazoles/farmacología , Virología/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...