Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1860(3): 607-17, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26655694

RESUMO

Replication of the HIV-1 virus requires reverse transcription of the viral RNA genome, a process that is specifically initiated by human tRNA3(Lys) packaged within the infectious virion. The primary binding site for the tRNA involves the 3' 18 nucleotides with an additional interaction between an adenine rich loop (A-loop) in the template and the anticodon stem-loop region of the tRNA3(Lys). The loop of the tRNA primer contains two hypermodified base residues and a pseudouridine that are required for a proper binding and activity. Here, we investigate the influence on the structure, dynamics and binding stability of the three modified residues (mnm(5)s(2)U34, t(6)A37 and Ψ39) using extensive molecular dynamics and Quantum Theory of Atoms in Molecules (QTAIM) analysis. Consistent with experiment, the results suggest that the three modified residues are required for faithful binding. Residues mnm(5)s(2)U34 and Ψ39 have a major influence in stabilizing the anticodon loop whereas mnm(5)s(2)U34 and t(6)A37 appear to stabilize the formation of the complex of tRNA3(Lys) with the HIV-1 A-loop.


Assuntos
Anticódon , Primers do DNA/química , HIV-1/genética , RNA de Transferência de Lisina/química , Elétrons
2.
J Chem Inf Model ; 54(6): 1758-72, 2014 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-24835734

RESUMO

Despite the many biological functions of RNA, very few drugs have been designed or found to target RNA. Here we report the results of molecular dynamics (MD) simulations and binding energy analyses on hepatitis C virus internal ribosome entry site (IRES) RNA in complex with highly charged 2-aminobenzimidazole inhibitors. Initial coordinates were taken from NMR and crystallography studies that had yielded different binding modes. During MD simulations, the RNA-inhibitor complex is stable in the crystal conformation but not in the NMR conformation. Additionally, we found that existing and standard MD trajectory postprocessing free energy methods, such as the MM-GBSA and MM-PBSA approaches available in AMBER, seem unsuitable to properly rank the binding energies of complexes between highly charged molecules. A better correlation with the experimental data was found using a rather simple binding enthalpy calculation based on the explicitly solvated potential energies. In anticipation of further growth in the use of small molecules to target RNA, we include results addressing the impact of charge assignment on docking, the structural role of magnesium in the IRES-inhibitor complex, the entropic contribution to binding energy, and simulations of a plausible scaffold design for new inhibitors.


Assuntos
Benzimidazóis/química , Benzimidazóis/farmacologia , Hepacivirus/metabolismo , Simulação de Dinâmica Molecular , RNA Viral/antagonistas & inibidores , Sequência de Bases , Hepacivirus/química , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Humanos , Simulação de Acoplamento Molecular , RNA Viral/química , RNA Viral/metabolismo , Termodinâmica
3.
J Biomol NMR ; 53(4): 321-39, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22714631

RESUMO

Restrained molecular dynamics simulations are a robust, though perhaps underused, tool for the end-stage refinement of biomolecular structures. We demonstrate their utility-using modern simulation protocols, optimized force fields, and inclusion of explicit solvent and mobile counterions-by re-investigating the solution structures of two RNA hairpins that had previously been refined using conventional techniques. The structures, both domain 5 group II intron ribozymes from yeast ai5γ and Pylaiella littoralis, share a nearly identical primary sequence yet the published 3D structures appear quite different. Relatively long restrained MD simulations using the original NMR restraint data identified the presence of a small set of violated distance restraints in one structure and a possibly incorrect trapped bulge nucleotide conformation in the other structure. The removal of problematic distance restraints and the addition of a heating step yielded representative ensembles with very similar 3D structures and much lower pairwise RMSD values. Analysis of ion density during the restrained simulations helped to explain chemical shift perturbation data published previously. These results suggest that restrained MD simulations, with proper caution, can be used to "update" older structures or aid in the refinement of new structures that lack sufficient experimental data to produce a high quality result. Notable cautions include the need for sufficient sampling, awareness of potential force field bias (such as small angle deviations with the current AMBER force fields), and a proper balance between the various restraint weights.


Assuntos
Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , RNA/química , Íntrons , Modelos Moleculares , Conformação de Ácido Nucleico , RNA Catalítico/química
4.
Antivir Chem Chemother ; 21(3): 117-28, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21233533

RESUMO

HCV infection is a significant human disease, leading to liver cirrhosis and cancer, and killing >10,000 people in the US annually. Translation of the viral RNA genome is initiated by ribosomal binding to a highly structured RNA element, the internal ribosomal entry site (IRES), which presents a novel target for therapeutic intervention. We will first discuss studies of oligonucleotide therapeutics targeting various regions of the 340-nucleotide IRES, many of which have effectively blocked IRES function in vitro and are active against virus replication in cell culture. Although low nanomolar potencies have been obtained for DNA- and RNA-based molecules, stability and drug delivery challenges remain to be addressed for these particular HCV compounds. Several classes of small molecule inhibitors have been identified from screening protocols or designed from established RNA therapeutic scaffolds. In particular, small molecule IRES inhibitors based on a benzimidazole scaffold bind specifically to the IRES, and inhibit viral replication in cell culture at micromolar concentrations with low toxicity. The structure of the RNA target in complex with a representative member of these small molecule inhibitors demonstrates that a large RNA conformational change occurs upon inhibitor binding. The RNA complex shows how the inhibitor alters the global RNA structure and provides a framework for structure-based drug design of novel HCV therapeutics.


Assuntos
Hepacivirus/genética , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/genética , RNA Ribossômico/genética , RNA Viral/genética , Humanos , Terapia de Alvo Molecular/métodos , RNA Ribossômico/efeitos dos fármacos , RNA Ribossômico/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
5.
Antiviral Res ; 89(1): 54-63, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21075143

RESUMO

Major progress has been made in developing infectious HCV cell culture systems and these systems have been useful in identifying novel HCV antivirals. However, more rapid and sensitive assays using infectious cell based HCV systems would facilitate the development of additional antivirals, including small molecules directed at unique targets such as the HCV RNA internal ribosomal entry site (IRES). We have found that the V3 region (28 aa) of NS5A of HCV JFH1 can be deleted from the genome with only modest effects on the titer of infectious virus produced in cell culture. Moreover, the V3 region can be replaced with the Renilla reniformis luciferase (Rluc) gene resulting in an infectious virus that stably expresses an NS5A-Rluc fusion protein. Infected cells cultured in 96-well plates provided a robust luciferase signal that accurately reflected the production of infectious virus. This infectious HCV reporter system was used to test the activity of three benzimidazole compounds that bind the HCV RNA IRES. Compounds in this chemical class of small molecules bind and alter the IRES RNA structure at low to sub-micromolar concentrations and interfere with viral replication. The current study shows that these compounds inhibit HCV replication in an infectious HCV cell culture system, defines their IC(50) in this system, and provides a platform for the rapid testing of next generation inhibitors.


Assuntos
Antivirais/farmacologia , Benzimidazóis/farmacologia , Hepacivirus/efeitos dos fármacos , Hepacivirus/crescimento & desenvolvimento , Biossíntese de Proteínas/efeitos dos fármacos , Animais , Fusão Gênica Artificial , Linhagem Celular , Genes Reporter , Hepatócitos/virologia , Humanos , Concentração Inibidora 50 , Luciferases de Renilla/genética , Luciferases de Renilla/metabolismo , Testes de Sensibilidade Microbiana/métodos , Renilla , Coloração e Rotulagem/métodos
6.
Proc Natl Acad Sci U S A ; 107(16): 7263-8, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20360559

RESUMO

Translation of the hepatitis C virus (HCV) RNA is initiated from a highly structured internal ribosomal entry site (IRES) in the 5' untranslated region (5' UTR) of the RNA genome. An important structural feature of the native RNA is an approximately 90 degrees helical bend localized to domain IIa that positions the apical loop of domain IIb of the IRES near the 40S ribosomal E-site to promote eIF2-GDP release, facilitating 80S ribosome assembly. We report here the NMR structure of a domain IIa construct in complex with a potent small-molecule inhibitor of HCV replication. Molecular dynamics refinement in explicit solvent and subsequent energetic analysis indicated that each inhibitor stereoisomer bound with comparable affinity and in an equivalent binding mode. The in silico analysis was substantiated by fluorescence-based assays showing that the relative binding free energies differed by only 0.7 kcal/mol. Binding of the inhibitor displaces key nucleotide residues within the bulge region, effecting a major conformational change that eliminates the bent RNA helical trajectory, providing a mechanism for the antiviral activity of this inhibitor class.


Assuntos
Hepacivirus/genética , RNA Viral/química , Sequência de Bases , Fator de Iniciação 2 em Eucariotos/química , Hepacivirus/metabolismo , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética/métodos , Conformação Molecular , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Ligação Proteica , Conformação Proteica , RNA Ribossômico/química , Solventes/química , Eletricidade Estática , Estereoisomerismo
7.
J Am Chem Soc ; 130(43): 14280-6, 2008 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-18831583

RESUMO

Disulfide-rich peptides represent a megadiverse group of natural products with very promising therapeutic potential. To accelerate their functional characterization, high-throughput chemical synthesis and folding methods are required, including efficient mapping of multiple disulfide bridges. Here, we describe a novel approach for such mapping and apply it to a three-disulfide-bridged conotoxin, mu-SxIIIA (from the venom of Conus striolatus), whose discovery is also reported here for the first time. Mu-SxIIIA was chemically synthesized with three cysteine residues labeled 100% with (15)N/(13)C, while the remaining three cysteine residues were incorporated using a mixture of 70%/30% unlabeled/labeled Fmoc-protected residues. After oxidative folding, the major product was analyzed by NMR spectroscopy. Sequence-specific resonance assignments for the isotope-enriched Cys residues were determined with 2D versions of standard triple-resonance ((1)H, (13)C, (15)N) NMR experiments and 2D [(13)C, (1)H] HSQC. Disulfide patterns were directly determined with cross-disulfide NOEs confirming that the oxidation product had the disulfide connectivities characteristic of mu-conotoxins. Mu-SxIIIA was found to be a potent blocker of the sodium channel subtype Na(V)1.4 (IC50 = 7 nM). These results suggest that differential incorporation of isotope-labeled cysteine residues is an efficient strategy to map disulfides and should facilitate the discovery and structure-function studies of many bioactive peptides.


Assuntos
Conotoxinas/química , Cisteína/química , Dissulfetos/química , Espectroscopia de Ressonância Magnética/métodos , Peptídeos Cíclicos/química , Animais , Caramujo Conus , Espectroscopia de Ressonância Magnética/normas , Oxirredução , Dobramento de Proteína , Padrões de Referência , Fatores de Tempo
8.
Nucleic Acids Res ; 35(14): 4704-14, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17604307

RESUMO

The optional Escherichia coli restriction tRNase PrrC represents a family of potential antiviral devices widespread among bacteria. PrrC comprises a functional C-domain of unknown structure and regulatory ABC/ATPase-like N-domain. The possible involvement of a C-domain sequence in tRNA(Lys) recognition was investigated using a matching end-protected 11-meric peptide. This mimic, termed here LARP (Lys-anticodon recognizing peptide) UV-cross-linked tRNA(Lys) anticodon stem-loop (ASL) analogs and inhibited their PrrC-catalyzed cleavage. Trimming LARP or introducing in it inactivating PrrC missense mutations impaired these activities. LARP appeared to mimic its matching protein sequence in ability to dimerize in parallel, as inferred from the following results. First, tethering Cys to the amino- or carboxy-end of LARP dramatically enhanced the ASL-cross-linking and PrrC-inhibiting activities under suitable redox conditions. Second, Cys-substitutions in a C-domain region containing the sequence corresponding to LARP elicited specific intersubunit cross-links. The parallel dimerization of PrrC's C-domains and expected head-to-tail dimerization of its N-domains further suggest that the NTPase and tRNA(Lys)-binding sites of PrrC arise during distinct assembly stages of its dimer of dimers form.


Assuntos
Proteínas de Escherichia coli/química , RNA de Transferência de Lisina/química , Ribonucleases/química , Sequência de Aminoácidos , Sítios de Ligação , Cisteína/química , Dimerização , Proteínas de Escherichia coli/metabolismo , Mimetismo Molecular , Dados de Sequência Molecular , Mutagênese , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Oxirredução , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Ribonucleases/metabolismo , Alinhamento de Sequência , Raios Ultravioleta
9.
Biochemistry ; 44(22): 8078-89, 2005 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-15924427

RESUMO

Previous nuclear magnetic resonance (NMR) studies of unmodified and pseudouridine39-modified tRNA(Lys) anticodon stem loops (ASLs) show that significant structural rearrangements must occur to attain a canonical anticodon loop conformation. The Escherichia coli tRNA(Lys) modifications mnm(5)s(2)U34 and t(6)A37 have indeed been shown to remodel the anticodon loop, although significant dynamic flexibility remains within the weakly stacked U35 and U36 anticodon residues. The present study examines the individual effects of mnm(5)s(2)U34, s(2)U34, t(6)A37, and Mg(2+) on tRNA(Lys) ASLs to decipher how the E. coli modifications accomplish the noncanonical to canonical structural transition. We also investigated the effects of the corresponding human tRNA(Lys,3) versions of the E. coli modifications, using NMR to analyze tRNA ASLs containing the nucleosides mcm(5)U34, mcm(5)s(2)U34, and ms(2)t(6)A37. The human wobble modification has a less dramatic loop remodeling effect, presumably because of the absence of a positive charge on the mcm(5) side chain. Nonspecific magnesium effects appear to play an important role in promoting anticodon stacking. Paradoxically, both t(6)A37 and ms(2)t(6)A37 actually decrease anticodon stacking compared to A37 by promoting U36 bulging. Rather than stack with U36, the t(6)A37 nucleotide in the free tRNAs is prepositioned to form a cross-strand stack with the first codon nucleotide as seen in the recent crystal structures of tRNA(Lys) ASLs bound to the 30S ribosomal subunit. Wobble modifications, t(6)A37, and magnesium each make unique contributions toward promoting canonical tRNA structure in the fundamentally dynamic tRNA(Lys)(UUU) anticodon.


Assuntos
Adenosina/análogos & derivados , Anticódon/química , Escherichia coli/genética , Conformação de Ácido Nucleico , RNA de Transferência de Lisina/química , Tionucleosídeos/química , Tiouridina/análogos & derivados , Adenosina/química , Pareamento de Bases , Sítios de Ligação/genética , Códon/química , Engenharia Genética , Humanos , Magnésio/química , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Pseudouridina/química , Ribossomos/química , Termodinâmica , Tiouridina/química
10.
Methods Mol Biol ; 288: 187-204, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15333904

RESUMO

The anticodon domain of lysine transfer ribonucleic acid (tRNA) is a model system for investigation of the structural and biochemical effects of nucleoside posttranscriptional modification. To enable detailed study of the biophysical and structural effects of hypermodified nucleosides, methods have been developed to synthesize RNA oligonucleotides containing the modified nucleosides found in lysine tRNA. We describe in detail the synthesis of protected phosphoramidites of the nucleosides methylaminomethyl-2-thiouridine (mnm5s2U), methylcarboxymethyl-2-thiouridine (mcm5s2U), and 2-thiomethyl-N-6-carbamoylthreonyl-adenosine (ms2t6A). We also describe methods for using these nucleoside phosphoramidite reagents to synthesize RNA oligonucleotides with modified nucleosides incorporated at the specific sequence locations corresponding to their positions in the native lysine tRNAs.


Assuntos
Nucleotídeos/química , RNA/química
11.
EMBO J ; 23(7): 1411-21, 2004 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-15029239

RESUMO

Ubiquitin (Ub) functions in many different biological pathways, where it typically interacts with proteins that contain modular Ub recognition domains. One such recognition domain is the Npl4 zinc finger (NZF), a compact zinc-binding module found in many proteins that function in Ub-dependent processes. We now report the solution structure of the NZF domain from Npl4 in complex with Ub. The structure reveals that three key NZF residues (13TF14/M25) surrounding the zinc coordination site bind the hydrophobic 'Ile44' surface of Ub. Mutations in the 13TF14/M25 motif inhibit Ub binding, and naturally occurring NZF domains that lack the motif do not bind Ub. However, substitution of the 13TF14/M25 motif into the nonbinding NZF domain from RanBP2 creates Ub-binding activity, demonstrating the versatility of the NZF scaffold. Finally, NZF mutations that inhibit Ub binding by the NZF domain of Vps36/ESCRT-II also inhibit sorting of ubiquitylated proteins into the yeast vacuole. Thus, the NZF is a versatile protein recognition domain that is used to bind ubiquitylated proteins during vacuolar protein sorting, and probably many other biological processes.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Estrutura Secundária de Proteína , Ubiquitina/metabolismo , Dedos de Zinco , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos , Mutação , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitina/química , Ubiquitina/genética
12.
Nucleic Acids Symp Ser (Oxf) ; (48): 263-4, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-17150579

RESUMO

Sequence placement of post-transcriptionally modified nucleosides in tRNA can be experimentally difficult, particularly in cases involving new or unexpected modifications or sequence sites. We describe a mass spectrometry-based approach to this problem, involving the following steps: crude isolations of one or several tRNAs by HPLC from an unfractionated tRNA mixture; digestion to oligonucleotide mixtures by RNase T1; analysis by combined HPLC/electrospray ionization-MS for recognition of modifications; and direct gas-phase sequencing of selected targets in the mixture by LC/MS/MS. Isoacceptor identity can be established in favorable cases when tRNA gene sequences are available.


Assuntos
Nucleotídeos/análise , RNA de Transferência/química , Análise de Sequência de RNA/métodos , Cromatografia Líquida de Alta Pressão , Espectrometria de Massas , Ribonuclease T1/metabolismo , Saccharomyces cerevisiae , Sulfolobus
13.
J Biol Chem ; 278(22): 20225-34, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12644454

RESUMO

Ubiquitylated proteins are directed into a large number of different cellular pathways through interactions with effector proteins that contain conserved ubiquitin binding motifs. Here, we report the solution structure and ubiquitin binding properties of one such motif, the Npl4 zinc finger or RanBP2/Nup358 zinc finger (NZF) domain. Npl4 NZF forms a compact module composed of four antiparallel beta-strands linked by three ordered loops. A single zinc ion is coordinated by four conserved cysteines from the first and third loops, which form two rubredoxin knuckles. Npl4 NZF binds specifically, but weakly, to free ubiquitin using a conserved 13TF14 dipeptide to interact with the "Ile-44" surface of ubiquitin. Our studies reveal the structure of this versatile class of protein binding domains and provide a means for identifying the subset of NZF domains likely to bind ubiquitin.


Assuntos
Proteínas/metabolismo , Ubiquitina/metabolismo , Dedos de Zinco , Animais , Microanálise por Sonda Eletrônica , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Conformação Proteica , Proteínas/química , Ratos , Zinco/metabolismo
14.
J Am Chem Soc ; 124(48): 14302-3, 2002 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-12452693

RESUMO

The HIV transcription initiation complex involves a putative interaction between the primer tRNA anticodon and a conserved A-rich loop in the HIV genome. Surface plasmon resonance was used to demonstrate that the hypermodified nucleosides in the tRNA anticodon stem loop (ASL) stabilize RNA-RNA interactions in a model for the anticodon/A-loop complex. tRNA ASL hairpins with the modifications of Escherchia coli tRNALys and human tRNALys,3 each form stable complexes. Partially modified tRNA ASLs bind the A-loop hairpin with lesser affinity, and it was found that the modifications of the bacterial and mammalian tRNAs make distinct contributions toward stabilizing the RNA complex. One model for the anticodon/A-loop RNA complex that is consistent with the known modification effects on tRNA structure and function is that of complementary tRNAs, as seen for the published crystal structure of tRNAAsp.


Assuntos
Adenosina/análogos & derivados , HIV-1/genética , Nucleosídeos/química , RNA de Transferência de Lisina/química , RNA Viral/química , Tiouridina/análogos & derivados , Adenosina/química , Anticódon/química , Anticódon/genética , Anticódon/metabolismo , Cinética , Conformação de Ácido Nucleico , Nucleosídeos/síntese química , Nucleosídeos/metabolismo , Compostos Organofosforados/síntese química , Compostos Organofosforados/química , RNA de Transferência de Lisina/genética , RNA de Transferência de Lisina/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Ressonância de Plasmônio de Superfície , Tiouridina/química
15.
Nat Struct Biol ; 9(11): 812-7, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12379843

RESUMO

The structural proteins of HIV and Ebola display PTAP peptide motifs (termed 'late domains') that recruit the human protein Tsg101 to facilitate virus budding. Here we present the solution structure of the UEV (ubiquitin E2 variant) binding domain of Tsg101 in complex with a PTAP peptide that spans the late domain of HIV-1 p6(Gag). The UEV domain of Tsg101 resembles E2 ubiquitin-conjugating enzymes, and the PTAP peptide binds in a bifurcated groove above the vestigial enzyme active site. Each PTAP residue makes important contacts, and the Ala 9-Pro 10 dipeptide binds in a deep pocket of the UEV domain that resembles the X-Pro binding pockets of SH3 and WW domains. The structure reveals the molecular basis of HIV PTAP late domain function and represents an attractive starting point for the design of novel inhibitors of virus budding.


Assuntos
Proteínas de Ligação a DNA/química , Produtos do Gene gag/química , HIV-1/química , Fatores de Transcrição/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Aminoácidos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte , Produtos do Gene gag/metabolismo , HIV-1/metabolismo , Substâncias Macromoleculares , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Fatores de Transcrição/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana
16.
J Org Chem ; 67(15): 5352-8, 2002 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-12126427

RESUMO

The synthesis of a protected form of the hypermodified nucleoside, N-[(9-beta-D-ribofuranosyl-2-methylthiopurin-6-yl)carbamoyl]threonine, (ms2t6A) is reported. The hypermodified nucleoside was subsequently elaborated to the protected nucleoside phosphophoramidite using a protecting group strategy compatible with standard RNA oligonucleotide chemistry. The phosphoramidite reagent was then used to synthesize the 17-nucleotide RNA hairpin having the sequence of the anticodon stem-loop (ASL) domain of human tRNA(Lys,3), the primer for HIV-1 reverse transcriptase. Introduction of the modification at position 37 of the tRNA ASL modestly decreases the thermodynamic stability of the RNA hairpin as has been seen previously for the prokaryotic t6A nucleoside lacking the 2-methylthio substituent. 2D NOESY NMR spectra of the ms2t6A containing tRNA ASL indicate that the threonyl side chain adopts a conformation similar to that seen in the solution structure of the analogous t6A containing E. coli tRNA(Lys), despite the presence of the bulky methylthio group. This synthetic approach allows for site-specific incorporation of the hypermodified nucleoside and should facilitate future studies directed at understanding the roles of nucleoside modification in modulating the stability and specificity of biologically important RNA-RNA interactions. Our synthesis of the ms2t6A containing RNAs demonstrates that this methodology is suitable for obtaining quantities of RNA required for structural studies of the HIV primer tRNA.


Assuntos
Anticódon/química , Nucleosídeos/síntese química , RNA de Transferência de Lisina/síntese química , RNA/química , RNA/síntese química , Tionucleosídeos/síntese química , Catálise , Dicroísmo Circular , Transcriptase Reversa do HIV/química , Humanos , Modelos Moleculares , Estrutura Molecular , Conformação de Ácido Nucleico , RNA de Transferência de Lisina/química , Espectrofotometria Ultravioleta , Tionucleosídeos/química
17.
EMBO J ; 21(10): 2397-406, 2002 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12006492

RESUMO

Human Tsg101 plays key roles in HIV budding and in cellular vacuolar protein sorting (VPS). In performing these functions, Tsg101 binds both ubiquitin (Ub) and the PTAP tetrapeptide 'late domain' motif located within the viral Gag protein. These interactions are mediated by the N-terminal domain of Tsg101, which belongs to the catalytically inactive ubiquitin E2 variant (UEV) family. We now report the structure of Tsg101 UEV and chemical shift mapping of the Ub and PTAP binding sites. Tsg101 UEV resembles canonical E2 ubiquitin conjugating enzymes, but has an additional N-terminal helix, an extended beta-hairpin that links strands 1 and 2, and lacks the two C-terminal helices normally found in E2 enzymes. PTAP-containing peptides bind in a hydrophobic cleft exposed by the absence of the C-terminal helices, whereas ubiquitin binds in a novel site surrounding the beta-hairpin. These studies provide a structural framework for understanding how Tsg101 mediates the protein-protein interactions required for HIV budding and VPS.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Complexos Endossomais de Distribuição Requeridos para Transporte , HIV/fisiologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Conformação Proteica , Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência , Ubiquitina/metabolismo , Dedos de Zinco
18.
J Biol Chem ; 277(6): 3836-41, 2002 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-11723135

RESUMO

The bacterial tRNA(Lys)-specific PrrC-anticodon nuclease efficiently cleaved an anticodon stem-loop (ASL) oligoribonucleotide containing the natural modified bases, suggesting this region harbors the specificity determinants. Assays of ASL analogs indicated that the 6-threonylcarbamoyl adenosine modification (t(6)A37) enhances the reactivity. The side chain of the modified wobble base 5-methylaminomethyl-2-thiouridine (mnm(5)s(2)U34) has a weaker positive effect depending on the context of other modifications. The s(2)U34 modification apparently has none and the pseudouridine (psi39) was inhibitory in most modification contexts. GC-rich but not IC-rich stems abolished the activity. Correlating the reported structural effects of the base modifications with their effects on anticodon nuclease activity suggests preference for substrates where the anticodon nucleotides assume a stacked A-RNA conformation and base pairing interactions in the stem are destabilized. Moreover, the proposal that PrrC residue Asp(287) contacts mnm(5)s(2)U34 was reinforced by the observations that the mammalian tRNA(Lys-3) wobble base 5-methoxycarbonyl methyl-2-thiouridine (mcm(5)s(2)U) is inhibitory and that the D287H mutant favors tRNA(Lys-3) over Escherichia coli tRNA(Lys). The detection of this mutation and ability of PrrC to cleave the isolated ASL suggest that anticodon nuclease may be used to cleave tRNA(Lys-3) primer molecules annealed to the genomic RNA template of the human immunodeficiency virus.


Assuntos
Conformação de Ácido Nucleico , RNA de Transferência de Lisina/química , Ribonucleases/metabolismo , Cinética , Mutação , RNA de Transferência de Lisina/genética , RNA de Transferência de Lisina/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...