Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Virol ; 96(1): e0134921, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34643428

RESUMEN

HIV-1 virion production is driven by Gag and Gag-Pol (GP) proteins, with Gag forming the bulk of the capsid and driving budding, while GP binds Gag to deliver the essential virion enzymes protease, reverse transcriptase, and integrase. Virion GP levels are traditionally thought to reflect the relative abundances of GP and Gag in cells (∼1:20), dictated by the frequency of a -1 programmed ribosomal frameshifting (PRF) event occurring in gag-pol mRNAs. Here, we exploited a panel of PRF mutant viruses to show that mechanisms in addition to PRF regulate GP incorporation into virions. First, we show that GP is enriched ∼3-fold in virions relative to cells, with viral infectivity being better maintained at subphysiological levels of GP than when GP levels are too high. Second, we report that GP is more efficiently incorporated into virions when Gag and GP are synthesized in cis (i.e., from the same gag-pol mRNA) than in trans, suggesting that Gag/GP translation and assembly are spatially coupled processes. Third, we show that, surprisingly, virions exhibit a strong upper limit to trans-delivered GP incorporation; an adaptation that appears to allow the virus to temper defects to GP/Gag cleavage that may negatively impact reverse transcription. Taking these results together, we propose a "weighted Goldilocks" scenario for HIV-1 GP incorporation, wherein combined mechanisms of GP enrichment and exclusion buffer virion infectivity over a broad range of local GP concentrations. These results provide new insights into the HIV-1 virion assembly pathway relevant to the anticipated efficacy of PRF-targeted antiviral strategies. IMPORTANCE HIV-1 infectivity requires incorporation of the Gag-Pol (GP) precursor polyprotein into virions during the process of virus particle assembly. Mechanisms dictating GP incorporation into assembling virions are poorly defined, with GP levels in virions traditionally thought to solely reflect relative levels of Gag and GP expressed in cells, dictated by the frequency of a -1 programmed ribosomal frameshifting (PRF) event that occurs in gag-pol mRNAs. Herein, we provide experimental support for a "weighted Goldilocks" scenario for GP incorporation, wherein the virus exploits both random and nonrandom mechanisms to buffer infectivity over a wide range of GP expression levels. These mechanistic data are relevant to ongoing efforts to develop antiviral strategies targeting PRF frequency and/or HIV-1 virion maturation.


Asunto(s)
Sistema de Lectura Ribosómico , Regulación Viral de la Expresión Génica , Infecciones por VIH/virología , VIH-1/fisiología , Ensamble de Virus , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , VIH-1/efectos de los fármacos , Humanos , Secuencias Invertidas Repetidas , Modelos Biológicos , Conformación de Ácido Nucleico , Estabilidad del ARN , ARN Viral/química , ARN Viral/genética , Virión , Replicación Viral
2.
Nucleic Acids Res ; 46(21): 11488-11501, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30215753

RESUMEN

Post-transcriptional modification of snRNA is central to spliceosome function. Usb1 is an exoribonuclease that shortens the oligo-uridine tail of U6 snRNA, resulting in a terminal 2',3' cyclic phosphate group in most eukaryotes, including humans. Loss of function mutations in human Usb1 cause the rare disorder poikiloderma with neutropenia (PN), and result in U6 snRNAs with elongated 3' ends that are aberrantly adenylated. Here, we show that human Usb1 removes 3' adenosines with 20-fold greater efficiency than uridines, which explains the presence of adenylated U6 snRNAs in cells lacking Usb1. We determined three high-resolution co-crystal structures of Usb1: wild-type Usb1 bound to the substrate analog adenosine 5'-monophosphate, and an inactive mutant bound to RNAs with a 3' terminal adenosine and uridine. These structures, along with QM/MM MD simulations of the catalytic mechanism, illuminate the molecular basis for preferential deadenylation of U6 snRNA. The extent of Usb1 processing is influenced by the secondary structure of U6 snRNA.


Asunto(s)
Adenosina/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , ARN Nuclear Pequeño/metabolismo , Uridina/metabolismo , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Secuencia de Bases , Cristalografía por Rayos X , Predisposición Genética a la Enfermedad/genética , Humanos , Modelos Moleculares , Mutación , Neutropenia/genética , Neutropenia/metabolismo , Conformación de Ácido Nucleico , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/genética , Unión Proteica , Conformación Proteica , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/genética , Anomalías Cutáneas/genética , Anomalías Cutáneas/metabolismo , Especificidad por Sustrato
3.
J Virol ; 90(15): 6906-6917, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27194769

RESUMEN

UNLABELLED: Human immunodeficiency virus (HIV) replication is strongly dependent upon a programmed ribosomal frameshift. Here we investigate the relationships between the thermodynamic stability of the HIV type 1 (HIV-1) RNA frameshift site stem-loop, frameshift efficiency, and infectivity, using pseudotyped HIV-1 and HEK293T cells. The data reveal a strong correlation between frameshift efficiency and local, but not overall, RNA thermodynamic stability. Mutations that modestly increase the local stability of the frameshift site RNA stem-loop structure increase frameshift efficiency 2-fold to 3-fold in cells. Thus, frameshift efficiency is determined by the strength of the thermodynamic barrier encountered by the ribosome. These data agree with previous in vitro measurements, suggesting that there are no virus- or host-specific factors that modulate frameshifting. The data also indicate that there are no sequence-specific requirements for the frameshift site stem-loop. A linear correlation between Gag-polymerase (Gag-Pol) levels in cells and levels in virions supports the idea of a stochastic virion assembly mechanism. We further demonstrate that the surrounding genomic RNA secondary structure influences frameshift efficiency and that a mutation that commonly arises in response to protease inhibitor therapy creates a functional but inefficient secondary slippery site. Finally, HIV-1 mutants with enhanced frameshift efficiencies are significantly less infectious, suggesting that compounds that increase frameshift efficiency by as little as 2-fold may be effective at suppressing HIV-1 replication. IMPORTANCE: HIV, like many retroviruses, utilizes a -1 programmed ribosomal frameshift to generate viral enzymes in the form of a Gag-Pol polyprotein precursor. Thus, frameshifting is essential for viral replication. Here, we utilized a panel of mutant HIV strains to demonstrate that in cells, frameshifting efficiency is correlated with the stability of the local thermodynamic barrier to ribosomal translocation. Increasing the stability of the frameshift site RNA increases the frameshift efficiency 2-fold to 3-fold. Mutant viruses with increased frameshift efficiencies have significantly reduced infectivity. These data suggest that this effect might be exploited in the development of novel antiviral strategies.


Asunto(s)
Mutación del Sistema de Lectura/genética , Sistema de Lectura Ribosómico/genética , Proteínas de Fusión gag-pol/metabolismo , Infecciones por VIH/virología , VIH-1/genética , ARN Viral/genética , Virión/fisiología , Emparejamiento Base , Secuencia de Bases , Regulación Viral de la Expresión Génica , Células HEK293 , Infecciones por VIH/genética , VIH-1/química , VIH-1/metabolismo , Humanos , Conformación de Ácido Nucleico , Estabilidad del ARN , ARN Viral/química , ARN Viral/metabolismo , Ensamble de Virus , Replicación Viral
4.
ACS Chem Biol ; 11(1): 88-94, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26496521

RESUMEN

Human Immunodeficiency Virus (HIV) type 1 uses a -1 programmed ribosomal frameshift (-1 PRF) event to translate its enzymes from the same transcript used to encode the virus' structural proteins. The frequency of this event is highly regulated, and significant deviation from the normal 5-10% frequency has been demonstrated to decrease viral infectivity. Frameshifting is primarily regulated by the Frameshift Stimulatory Signal RNA (FSS-RNA), a thermodynamically stable, highly conserved stem loop that has been proposed as a therapeutic target. We describe the design, synthesis, and testing of a series of N-methyl peptides able to bind the HIV-1 FSS RNA stem loop with low nanomolar affinity and high selectivity. Surface plasmon resonance (SPR) data indicates increased affinity is a reflection of a substantially enhanced on rate. Compounds readily penetrate cell membranes and inhibit HIV infectivity in a pseudotyped virus assay. Viral infectivity inhibition correlates with compound-dependent changes in the ratios of Gag and Gag-Pol in virus particles. As the first compounds with both single digit nanomolar affinities for the FSS RNA and an ability to inhibit HIV in cells, these studies support the use of N-methylation for enhancing the affinity, selectivity, and bioactivity of RNA-binding peptides.


Asunto(s)
Sistema de Lectura Ribosómico/fisiología , VIH-1/química , ARN Viral/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Secuencia de Bases , Sistema de Lectura Ribosómico/genética , Células HEK293 , VIH-1/efectos de los fármacos , Humanos , Metilación , Datos de Secuencia Molecular , Estructura Molecular , Péptidos/síntesis química , Péptidos/metabolismo , Unión Proteica , ARN Viral/química
5.
Proc Natl Acad Sci U S A ; 112(47): E6446-55, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26554019

RESUMEN

The dicistrovirus intergenic region internal ribosome entry site (IRES) adopts a triple-pseudoknotted RNA structure and occupies the core ribosomal E, P, and A sites to directly recruit the ribosome and initiate translation at a non-AUG codon. A subset of dicistrovirus IRESs directs translation in the 0 and +1 frames to produce the viral structural proteins and a +1 overlapping open reading frame called ORFx, respectively. Here we show that specific mutations of two unpaired adenosines located at the core of the three-helical junction of the honey bee dicistrovirus Israeli acute paralysis virus (IAPV) IRES PKI domain can uncouple 0 and +1 frame translation, suggesting that the structure adopts distinct conformations that contribute to 0 or +1 frame translation. Using a reconstituted translation system, we show that ribosomes assembled on mutant IRESs that direct exclusive 0 or +1 frame translation lack reading frame fidelity. Finally, a nuclear magnetic resonance/small-angle X-ray scattering hybrid approach reveals that the PKI domain of the IAPV IRES adopts an RNA structure that resembles a complete tRNA. The tRNA shape-mimicry enables the viral IRES to gain access to the ribosome tRNA-binding sites and form intermolecular contacts with the ribosome that are necessary for initiating IRES translation in a specific reading frame.


Asunto(s)
Dicistroviridae/genética , Sitios Internos de Entrada al Ribosoma/genética , Sistemas de Lectura Abierta/genética , Biosíntesis de Proteínas , ARN de Transferencia/química , ARN de Transferencia/genética , Animales , Secuencia de Bases , Bovinos , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Conformación de Ácido Nucleico , Factores de Terminación de Péptidos , Ribosomas/metabolismo , Dispersión del Ángulo Pequeño , Difracción de Rayos X
6.
Proc Natl Acad Sci U S A ; 112(14): 4292-7, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25831501

RESUMEN

RecQ helicases unwind remarkably diverse DNA structures as key components of many cellular processes. How RecQ enzymes accommodate different substrates in a unified mechanism that couples ATP hydrolysis to DNA unwinding is unknown. Here, the X-ray crystal structure of the Cronobacter sakazakii RecQ catalytic core domain bound to duplex DNA with a 3' single-stranded extension identifies two DNA-dependent conformational rearrangements: a winged-helix domain pivots ∼90° to close onto duplex DNA, and a conserved aromatic-rich loop is remodeled to bind ssDNA. These changes coincide with a restructuring of the RecQ ATPase active site that positions catalytic residues for ATP hydrolysis. Complex formation also induces a tight bend in the DNA and melts a portion of the duplex. This bending, coupled with translocation, could provide RecQ with a mechanism for unwinding duplex and other DNA structures.


Asunto(s)
Bacterias/enzimología , Cronobacter/enzimología , ADN/química , RecQ Helicasas/química , Adenosina Trifosfato/química , Anisotropía , Sitios de Unión , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Análisis Mutacional de ADN , ADN de Cadena Simple/química , Escherichia coli/enzimología , Genoma Bacteriano , Hidrólisis , Unión Proteica
7.
Genes Dev ; 26(23): 2634-46, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23207918

RESUMEN

Escherichia coli DksA is a transcription factor that binds to RNA polymerase (RNAP) without binding to DNA, destabilizing RNAP-promoter interactions, sensitizing RNAP to the global regulator ppGpp, and regulating transcription of several hundred target genes, including those encoding rRNA. Previously, we described promoter sequences and kinetic properties that account for DksA's promoter specificity, but how DksA exerts its effects on RNAP has remained unclear. To better understand DksA's mechanism of action, we incorporated benzoyl-phenylalanine at specific positions in DksA and mapped its cross-links to RNAP, constraining computational docking of the two proteins. The resulting evidence-based model of the DksA-RNAP complex as well as additional genetic and biochemical approaches confirmed that DksA binds to the RNAP secondary channel, defined the orientation of DksA in the channel, and predicted a network of DksA interactions with RNAP that includes the rim helices and the mobile trigger loop (TL) domain. Engineered cysteine substitutions in the TL and DksA coiled-coil tip generated a disulfide bond between them, and the interacting residues were absolutely required for DksA function. We suggest that DksA traps the TL in a conformation that destabilizes promoter complexes, an interaction explaining the requirement for the DksA tip and its effects on transcription.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Regulación Bacteriana de la Expresión Génica , ARN Polimerasas Dirigidas por ADN/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Modelos Moleculares , Fenilalanina/análogos & derivados , Fenilalanina/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo
8.
ACS Chem Biol ; 1(6): 341-5, 2006 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-17163768

RESUMEN

Riboswitches are metabolite-sensing RNA structures that have been discovered in regulatory regions of messenger RNA (mRNA). They have the remarkable ability to shut off the transcription or translation of their own mRNAs in response to binding a specific metabolite. In other words, riboswitches regulate their own genes using RNA instead of protein. Three new crystal structures reveal how S-adenosylmethionine and thiamine pyrophosphate riboswitches accomplish this task.


Asunto(s)
Aptámeros de Nucleótidos/química , Regulación de la Expresión Génica/genética , ARN/química , S-Adenosilmetionina/química , Tiamina Pirofosfato/química , Tiamina Pirofosfato/fisiología , Animales , Humanos , ARN/fisiología , S-Adenosilmetionina/genética , S-Adenosilmetionina/fisiología , Tiamina Pirofosfato/genética
9.
J Mol Biol ; 351(2): 371-82, 2005 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-16002091

RESUMEN

Tertiary interactions are critical for proper RNA folding and ribozyme catalysis. RNA tertiary structure is often condensed through long-range helical packing interactions mediated by loop-receptor motifs. RNA structures displaying helical packing by loop-receptor interactions have been solved by X-ray crystallography, but not by NMR. Here, we report the NMR structure of a 30 kDa GAAA tetraloop-receptor RNA complex. In order to stabilize the complex, we used a modular design in which the RNA was engineered to form a homodimer, with each subunit containing a GAAA tetraloop phased one helical turn apart from its cognate 11-nucleotide receptor domain. The structure determination utilized specific isotopic labeling patterns (2H, 13C and 15N) and refinement against residual dipolar couplings. We observe a unique and highly unusual chemical shift pattern for an adenosine platform interaction that reveals a spectroscopic fingerprint for this motif. The structure of the GAAA tetraloop-receptor interaction is well defined solely from experimental NMR data, shows minor deviations from previously solved crystal structures, and verifies the previously inferred hydrogen bonding patterns within this motif. This work demonstrates the feasibility of using engineered homodimers as modular systems for the determination of RNA tertiary interactions by NMR.


Asunto(s)
Conformación de Ácido Nucleico , ARN/química , Secuencia de Bases , Cristalografía por Rayos X , Dimerización , Electroforesis en Gel de Poliacrilamida , Enlace de Hidrógeno , Cinética , Magnesio/química , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Protones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA