Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Photochem Photobiol B ; 100(1): 19-29, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20570168

ABSTRACT

Aryl azide-mediated photo cross-linking has been widely used to obtain structural features in biological systems, even though the reactive species generated upon photolysis in aqueous solution have not been well characterized. We have established a mechanistic framework for the formation of adducts between photoactivated 5-azido-2-nitrobenzoyl reagents and protein functional groups. Photolysis of the aryl azide tethered to biotin via an amide linkage yields a cross-link with streptavidin. The ability of the pre-irradiated reagent to form a similar cross-link indicates that it is the long-lived reactive intermediate that contributes to the cross-link formation. The reactive intermediate forms an adduct with tryptophan. The sequence of the labeled peptide is found to be GlyTrp(*)ThrValAlaTrp(*)LysAsn, corresponding to residues 74-81 of the streptavidin sequence, where Trp(*) designates the modified Trp-75 and Trp-79. A peak at m/z 1455.1 corresponding to the calculated [M(peptide)+aryl nitrene+2O](+) molecular ion value has been observed for the labeled peptide. Product structure identification experiments support the assignment that the long-lived reactive intermediate is a p-nitro-N-arylhydroxylamine, which undergoes a number of transformations in aqueous solution leading to nitroso derivatives. A plausible mechanism of the interaction between tryptophan and nitroso compound is discussed.


Subject(s)
Azides/chemistry , Cross-Linking Reagents/chemistry , Nitroso Compounds/chemistry , Streptavidin/chemistry , Affinity Labels/chemistry , Amino Acid Sequence , Biotin/chemistry , Photolysis , Streptavidin/metabolism , Tryptophan/chemistry
2.
Nucleic Acids Res ; 34(1): 42-52, 2006.
Article in English | MEDLINE | ID: mdl-16394022

ABSTRACT

Metal ions are essential for DNA polymerase and RNase H activities of HIV-1 reverse transcriptase (RT). RT studies are routinely performed at 6-8 mM Mg2+, despite the fact that the in vivo concentration might be as low as 0.2 mM. We studied the influence of MgCl2 and ATP, which likely binds a significant fraction of the magnesium pool in vivo, on the DNA polymerase and RNase H activities of HIV-1 RT, its inhibition by nucleoside RT inhibitors (NRTIs) and primer unblocking by AZT-resistant RT. At low Mg2+ concentration, reverse transcription of a natural template strongly increased despite a dramatically reduced intrinsic polymerase activity under such conditions. Low Mg2+ concentrations affected the RNA stability and indirectly decreased its degradation by the RNase H activity. The reduced RNA degradation prevented premature dissociation of the template and primer strands that otherwise generated dead-end DNA products. In addition, low Mg2+ dramatically decreased the incorporation of NRTIs into DNA and increased nucleotide excision by AZT-resistant RT. The latter effect is also most likely owing to the diminished cleavage of the RNA template. Thus, differences in the free Mg2+ concentration between different cell types or during the cell cycle might strongly affect HIV-1 replication and its inhibition.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/metabolism , Magnesium/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcription , Adenosine Triphosphate/pharmacology , DNA/biosynthesis , DNA Primers , DNA, Single-Stranded/biosynthesis , Drug Resistance, Viral , Nucleosides/pharmacology , Reverse Transcription/drug effects , Ribonuclease H/metabolism , Zidovudine/pharmacology
3.
J Mol Biol ; 354(1): 55-72, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16236319

ABSTRACT

The viral infectivity factor (Vif) protein of human immunodeficiency virus type 1 (HIV-1) is essential for viral replication in vivo. Packaging of Vif into viral particles is mediated by an interaction with viral genomic RNA and association with viral nucleoprotein complexes. Despite recent findings on the RNA-binding properties of Vif suggesting that Vif could be involved in retroviral assembly, no RNA sequence or structure specificity has been determined so far. To gain further insight into the mechanisms by which Vif might regulate viral replication, we studied the interactions of Vif with HIV-1 genomic RNA in vitro. Using extensive biochemical analysis, we have measured the affinity of recombinant Vif proteins for synthetic RNAs corresponding to various regions of the HIV-1 genome. We found that recombinant Vif proteins bind specifically to HIV-1 viral RNA fragments corresponding to the 5'-untranslated region (5'-UTR), gag and the 5' part of pol (K(d) between 45 nM and 65 nM). RNA encompassing nucleotides 1-497 or 499-996 of the HIV-1 genomic RNA bind 9+/-2 and 21+/-3 Vif molecules, respectively, and at least some of these proteins bind in a cooperative manner (Hill constant alpha(H) = 2.3). In contrast, RNAs corresponding to other parts of the HIV-1 genome or heterologous RNAs showed poor binding capacity and weak cooperativity (K(d) > 200 nM). Moreover, RNase T1 footprinting revealed a hierarchical binding of Vif, pointing to TAR and the poly(A) stem-loop structures as primary strong affinity targets, and downstream structures as secondary sites with moderate affinity. Taken together, our findings suggest that Vif may assist other proteins to maintain a correct folding of the genomic RNA in order to facilitate its packaging and further steps such as reverse transcription. Interestingly, our results suggest also that Vif could bind the viral RNA in order to protect it from the action of the antiviral factor APOBEC-3G/3F.


Subject(s)
5' Untranslated Regions/metabolism , Gene Products, vif/metabolism , HIV-1/genetics , HIV-1/metabolism , RNA, Viral/metabolism , 5' Untranslated Regions/chemistry , Base Sequence , Electrophoretic Mobility Shift Assay , HIV Long Terminal Repeat , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , RNA, Viral/chemistry , RNA-Binding Proteins/metabolism , Recombinant Proteins/metabolism , vif Gene Products, Human Immunodeficiency Virus
4.
Science ; 308(5718): 120-3, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15802605

ABSTRACT

The ribosome of Thermus thermophilus was cocrystallized with initiator transfer RNA (tRNA) and a structured messenger RNA (mRNA) carrying a translational operator. The path of the mRNA was defined at 5.5 angstroms resolution by comparing it with either the crystal structure of the same ribosomal complex lacking mRNA or with an unstructured mRNA. A precise ribosomal environment positions the operator stem-loop structure perpendicular to the surface of the ribosome on the platform of the 30S subunit. The binding of the operator and of the initiator tRNA occurs on the ribosome with an unoccupied tRNA exit site, which is expected for an initiation complex. The positioning of the regulatory domain of the operator relative to the ribosome elucidates the molecular mechanism by which the bound repressor switches off translation. Our data suggest a general way in which mRNA control elements must be placed on the ribosome to perform their regulatory task.


Subject(s)
Protein Biosynthesis , RNA, Bacterial/chemistry , RNA, Messenger/chemistry , Regulatory Sequences, Ribonucleic Acid , Repressor Proteins/metabolism , Ribosomes/metabolism , Thermus thermophilus/metabolism , Bacterial Proteins/metabolism , Base Pairing , Binding Sites , Crystallization , Crystallography, X-Ray , Fourier Analysis , Models, Molecular , Nucleic Acid Conformation , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/metabolism , RNA, Transfer, Met/chemistry , RNA, Transfer, Met/metabolism , Ribosomal Proteins/metabolism , Thermus thermophilus/genetics , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism
5.
EMBO J ; 24(4): 824-35, 2005 Feb 23.
Article in English | MEDLINE | ID: mdl-15678100

ABSTRACT

Staphylococcus aureus RNAIII is one of the largest regulatory RNAs, which controls several virulence genes encoding exoproteins and cell-wall-associated proteins. One of the RNAIII effects is the repression of spa gene (coding for the surface protein A) expression. Here, we show that spa repression occurs not only at the transcriptional level but also by RNAIII-mediated inhibition of translation and degradation of the stable spa mRNA by the double-strand-specific endoribonuclease III (RNase III). The 3' end domain of RNAIII, partially complementary to the 5' part of spa mRNA, efficiently anneals to spa mRNA through an initial loop-loop interaction. Although this annealing is sufficient to inhibit in vitro the formation of the translation initiation complex, the coordinated action of RNase III is essential in vivo to degrade the mRNA and irreversibly arrest translation. Our results further suggest that RNase III is recruited for targeting the paired RNAs. These findings add further complexity to the expression of the S. aureus virulon.


Subject(s)
Antigens, Bacterial/genetics , Gene Expression Regulation, Bacterial , RNA, Antisense/metabolism , RNA, Bacterial/metabolism , Ribonuclease III/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Base Sequence , Binding Sites , Molecular Sequence Data , Nucleic Acid Conformation , Protein Biosynthesis , RNA Stability , RNA, Antisense/chemistry , RNA, Antisense/genetics , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism , Staphylococcal Protein A/genetics , Staphylococcal Protein A/metabolism , Transcription, Genetic/genetics
6.
J Biol Chem ; 279(46): 48397-403, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15355993

ABSTRACT

With the increasing interest of RNAs in regulating a range of cell biological processes, very little is known about the structure of RNAs in tissue culture cells. We focused on the 5'-untranslated region of the human immunodeficiency virus type 1 RNA genome, a highly conserved RNA region, which contains structural domains that regulate key steps in the viral replication cycle. Up until now, structural information only came from in vitro studies. Here, we developed chemical modification assays to test nucleotide accessibility directly in infected cells and viral particles, thus circumventing possible biases and artifacts linked to in vitro assays. The secondary structure of the 5'-untranslated region in infected cells points to the existence of the various stem-loop motifs associated to distinct functions, proposed from in vitro probing, mutagenesis, and phylogeny. However, compared with in vitro data, subtle differences were observed in the dimerization initiation site hairpin, and none of the proposed long range interactions were observed between the functional domains. Moreover, no global RNA rearrangement was observed; structural differences between infected cells and viral particles were limited to the primer binding site, which became protected against chemical modification upon tRNA(3) (Lys) annealing in virions and to the main packaging signal. In addition, our data suggested that the genomic RNA could already dimerize in the cytoplasm of infected cells. Taken together, our results provided the first analysis of the dynamic of RNA structure of the human immunodeficiency virus type 1 RNA genome during virus assembly ex vivo.


Subject(s)
5' Untranslated Regions , HIV-1/genetics , Nucleic Acid Conformation , RNA, Viral/chemistry , Virion/genetics , Base Sequence , Cell Line , Dimerization , Genome, Viral , HIV-1/metabolism , Humans , Molecular Sequence Data , Virion/metabolism , Virus Replication
7.
J Biol Chem ; 279(34): 35923-31, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15194685

ABSTRACT

HIV-1 reverse transcription is initiated from a tRNA(3)(Lys) molecule annealed to the viral RNA at the primer binding site (PBS), but the structure of the initiation complex of reverse transcription remains controversial. Here, we performed in situ structural probing, as well as in vitro structural and functional studies, of the initiation complexes formed by highly divergent isolates (MAL and NL4.3/HXB2). Our results show that the structure of the initiation complex is not conserved. In MAL, and according to sequence analysis in 14% of HIV-1 isolates, formation of the initiation complex is accompanied by complex rearrangements of the viral RNA, and extensive interactions with tRNA(3)(Lys) are required for efficient initiation of reverse transcription. In NL4.3, HXB2, and most isolates, tRNA(3)(Lys) annealing minimally affects the viral RNA structure and no interaction outside the PBS is required for optimal initiation of reverse transcription. We suggest that in MAL, extensive interactions with tRNA(3)(Lys) are required to drive the structural rearrangements generating the structural elements ultimately recognized by reverse transcriptase. In NL4.3 and HXB2, these elements are already present in the viral RNA prior to tRNA(3)(Lys) annealing, thus explaining that extensive interactions with the primer are not required. Interestingly, such interactions are required in HXB2 mutants designed to use a non-cognate tRNA as primer (tRNA(His)). In the latter case, the extended interactions are required to counteract a negative contribution associate with the alternate primer.


Subject(s)
HIV-1/physiology , RNA, Transfer, Lys , Reverse Transcription , Base Sequence , HIV Infections/genetics , HIV Infections/virology , HIV-1/chemistry , Humans , Molecular Sequence Data , Molecular Structure , Nucleic Acid Conformation , RNA, Transfer, Lys/chemistry , RNA, Transfer, Lys/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Sequence Alignment , Transcription Factors/chemistry , Transcription Factors/genetics , Virus Replication/genetics
8.
RNA Biol ; 1(1): 66-73, 2004 May.
Article in English | MEDLINE | ID: mdl-17194931

ABSTRACT

Ribosomal protein S15 is highly conserved among prokaryotes. It plays a pivotal role in the assembly of the central domain of the small ribosomal subunit and regulates its own expression by a feedback mechanism at the translational level. The protein recognizes two RNA targets (rRNA and mRNA) that share only partial similarity. Its interaction with 16S rRNA has been fully characterized, while mRNA interactions and regulatory mechanisms have been extensively studied in E. coli and in T. thermophilus. Recently, we have characterized which aminoacids are involved in E. coli mRNA recognition, using an in vivo assay allowing to identify S15 mutations affecting the S15-mRNA interactions without altering 30S subunit assembly. Here, we address the following questions: Are common determinants used by S15 to recognize its rRNA and mRNA targets? What is the extent of molecular mimicry? Is the regulatory mechanism conserved? Our results indicate that specific recognition of mRNA and rRNA relies on both mimicry and site differentiation. They also highlight the high plasticity of RNA to adapt to evolutionary constraints.


Subject(s)
Mutation , Protein Biosynthesis , Ribosomal Proteins/physiology , Base Sequence , Escherichia coli/metabolism , Evolution, Molecular , Models, Genetic , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Messenger/chemistry , RNA, Ribosomal/chemistry , Ribosomal Proteins/chemistry , Ribosomes/chemistry , Thermus thermophilus/metabolism
9.
J Biol Chem ; 279(6): 4560-9, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14607826

ABSTRACT

Dimerization of bcd mRNA was shown to be important for the formation of ribonucleoprotein particles and their localization in Drosophila embryo. The cis-element responsible for dimerization is localized in a stem-loop domain (domain III) containing two essential complementary 6-nucleotide sequences in a hairpin loop (LIIIb) and an interior loop (LIIIa). Such an RNA element can potentially generate single or double "hand-by-arm" interactions leading to open and closed complexes, respectively. The former retains the possibility of forming multimers, whereas the latter does not. We showed previously that dimerization proceeds through a two-step mechanism, which includes a transition from the reversible initiation complex into a very stable one. Here we have addressed the nature of the initial interactions and the mechanism of transition. We engineered a series of different RNA fragments with the capacity to form defined open dimers, multimers, or closed dimers. We compared their thermodynamic and kinetic behavior and mapped nucleotides involved in intermolecular interactions by enzymatic and chemical footprinting experiments and chemical modification interference. Our results indicate that the initiation step leads to a reversible open dimer, involving a more limited number of intermolecular base pairs than expected. The two loops play distinct roles in this process, and the structure of loop IIIb is more constrained than that of loop IIIa. Thus, loop IIIa appears to be the driving element of the recognition process. The initial open dimer is then converted into a stable closed dimer, possibly through a kinetically controlled mechanism.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Homeodomain Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Animals , Base Sequence , Dimerization , Drosophila melanogaster/embryology , Kinetics , Molecular Sequence Data , Nucleic Acid Conformation , RNA Interference , RNA Stability , RNA, Messenger/chemistry , Thermodynamics
10.
Nucleic Acids Res ; 31(19): 5764-75, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14500840

ABSTRACT

Reverse transcription of HIV-1 RNA is initiated from the 3' end of a tRNA3Lys molecule annealed to the primer binding site (PBS). An additional interaction between the anticodon loop of tRNA3Lys and a viral A-rich loop is required for efficient initiation of reverse transcription of the HIV-1 MAL isolate. In the HIV-1 HXB2 isolate, simultaneous mutations of the PBS and the A-rich loop (mutant His-AC), but not of the PBS alone (mutant His) allows the virus to stably utilize tRNA(His) as primer. However, mutant His-AC selects additional mutations during cell culture, generating successively His-AC-GAC and His-AC-AT-GAC. Here, we wanted to establish direct relationships between the evolution of these mutants in cell culture, their efficiency in initiating reverse transcription and the structure of the primer/template complexes in vitro. The initiation of reverse transcription of His and His-AC RNAs was dramatically reduced. However, His-AC-GAC RNA, which incorporated three adaptative point mutations, was reverse transcribed more efficiently than the wild type RNA. Incorporation of two additional mutations decreased the efficiency of the initiation of reverse transcription, which remained at the wild type level. Structural probing showed that even though both His-AC and His-AC-GAC RNAs can potentially interact with the anticodon loop of tRNA(His), only the latter template formed a stable interaction. Thus, our results showed that the selection of adaptative mutations by HIV-1 mutants utilizing tRNA(His) as primer was initially dictated by the efficiency of the initiation of reverse transcription, which relied on the existence of a stable interaction between the mutated A-rich loop and the anticodon loop of tRNA(His).


Subject(s)
Gene Expression Regulation, Viral , HIV-1/genetics , RNA, Transfer, His/metabolism , RNA, Viral/biosynthesis , Transcription Initiation Site , Transcription, Genetic , Base Sequence , DNA, Viral/biosynthesis , HIV Reverse Transcriptase/metabolism , HIV-1/metabolism , Kinetics , Macromolecular Substances , Molecular Sequence Data , Mutation , RNA Probes , RNA Processing, Post-Transcriptional , RNA, Viral/genetics , Sequence Alignment , Structure-Activity Relationship , Templates, Genetic
11.
Biol Cell ; 95(3-4): 221-8, 2003.
Article in English | MEDLINE | ID: mdl-12867085

ABSTRACT

The loss of the fragile X mental retardation protein (FMRP) is responsible for the most common cause of inherited mental retardation called the fragile X syndrome. FMRP is suspected to participate in the synaptic plasticity of neurons by acting on posttranscriptional control of gene expression. FMRP is an RNA binding protein that associates with mRNAs together with other proteins to form large ribonucleoprotein complexes. These complexes are proposed to participate in the transport, localization and translation of target mRNAs. Progress has been made recently in the identification of the mRNAs and the proteins present in these complexes and a possible connection with the micro-RNA dependent regulatory pathway has been established.


Subject(s)
Fragile X Syndrome/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Female , Fragile X Mental Retardation Protein , Fragile X Syndrome/metabolism , Humans , Macromolecular Substances , Male , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , Protein Biosynthesis/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Synaptic Transmission/genetics
12.
Biochimie ; 85(5): 521-5, 2003 May.
Article in English | MEDLINE | ID: mdl-12763311

ABSTRACT

HIV-1 utilizes cellular tRNA(3)(Lys) to prime the initiation of reverse transcription. The selective incorporation of cytoplasmic tRNA(3)(Lys) into HIV-1 particles was recently shown to involve the lysyl-tRNA synthetase, and hence, the encapsidated tRNA(3)(Lys) is likely to be aminoacylated. Here, we tested the effect of aminoacylation on the initiation of reverse transcription. We show that HIV-1 reverse transcriptase is unable to extend lysyl-tRNA(3)(Lys). In addition, the viral polymerase does not significantly enhance the rate of tRNA deacylation, in contrast with previous studies on avian retroviruses. Thus, aminoacylation of the primer tRNA might prevent the initiation of HIV-1 reverse transcription from taking place before viral budding and maturation.


Subject(s)
Acylation , HIV-1/physiology , RNA, Transfer, Lys/chemistry , Transcription, Genetic/physiology , Acetyltransferases/metabolism , Animals , Cattle , HIV Reverse Transcriptase/pharmacology , RNA/genetics , RNA, Transfer, Lys/drug effects , Transcription, Genetic/drug effects , Virus Assembly
13.
EMBO J ; 22(8): 1898-908, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682022

ABSTRACT

The 16S rRNA-binding ribosomal protein S15 is a key component in the assembly of the small ribosomal subunit in bacteria. We have shown that S15 from the extreme thermophile Thermus thermophilus represses the translation of its own mRNA in vitro, by interacting with the leader segment of its mRNA. The S15 mRNA-binding site was characterized by footprinting experiments, deletion analysis and site-directed mutagenesis. S15 binding triggers a conformational rearrangement of its mRNA into a fold that mimics the conserved three-way junction of the S15 rRNA-binding site. This conformational change masks the ribosome entry site, as demonstrated by direct competition between the ribosomal subunit and S15 for mRNA binding. A comparison of the T.thermophilus and Escherichia coli regulation systems reveals that the two regulatory mRNA targets do not share any similarity and that the mechanisms of translational inhibition are different. Our results highlight an astonishing plasticity of mRNA in its ability to adapt to evolutionary constraints, that contrasts with the extreme conservation of the rRNA-binding site.


Subject(s)
Nucleic Acid Conformation , Protein Biosynthesis , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Ribosomal Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Protein Binding , Protein Footprinting , RNA, Messenger/chemistry , Repressor Proteins/genetics , Ribosomal Proteins/genetics , Ribosomes/metabolism , Thermus thermophilus/genetics , Thermus thermophilus/metabolism
14.
Nucleic Acids Res ; 31(3): 850-9, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12560480

ABSTRACT

Reverse transcription of HIV-1 RNA is primed by a tRNA3(Lys) molecule bound at the primer binding site (PBS). Complex intermolecular interactions were proposed between tRNA3(Lys) and the RNA of the HIV-1 Mal isolate. Recently, an alternative interaction was proposed between the TPsiC stem of tRNA3(Lys) and a primer activation signal (PAS) of the Lai and Hxb2 RNAs, suggesting major structural variations in the reverse transcription complex of different HIV-1 strains. Here, we analyzed mutants of the Hxb2 RNA that prevent the interaction between the PAS and tRNA3(Lys) or/and a complementary sequence in the viral RNA. We compared the kinetics of reverse transcription of the wild type and mutant Hxb2 RNAs, using either tRNA3(Lys) or an 18mer oligoribonucleotide complementary to the PBS, which cannot interact with the PAS, as primers. We also used chemical probing to test the structure of the mutant and wild type RNAs, as well as the complex formed between the later RNA and tRNA3(Lys). These experiments, together with the analysis of long term replication data of mutant viruses obtained by C. Morrow and coworkers (Birmingham, USA) that use alternate tRNAs as primers, strongly suggest that the interaction between the Hxb2 PAS and tRNA3(Lys) does not exist. Instead, the effects of the vRNA mutations on reverse transcription seem to be linked to incorrect folding of the mutant RNAs.


Subject(s)
Gene Expression Regulation, Viral , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , RNA, Transfer, Amino Acyl/metabolism , Transcription, Genetic , Base Sequence , Binding Sites , DNA Primers , DNA, Viral/biosynthesis , Kinetics , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Oligoribonucleotides , RNA, Transfer, Amino Acyl/chemistry , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism
15.
Mol Microbiol ; 47(4): 961-74, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12581352

ABSTRACT

In addition to its role in tRNA aminoacylation, Escherichia coli threonyl-tRNA synthetase is a regulatory protein which binds a site, called the operator, located in the leader of its own mRNA and inhibits translational initiation by competing with ribosome binding. This work shows that the two essential steps of regulation, operator recognition and inhibition of ribosome binding, are performed by different domains of the protein. The catalytic and the C-terminal domain of the protein are involved in binding the two anticodon arm-like structures in the operator whereas the N-terminal domain of the enzyme is responsible for the competition with the ribosome. This is the first demonstration of a modular structure for a translational repressor and is reminiscent of that of transcriptional regulators. The mimicry between the operator and tRNA, suspected on the basis of previous experiments, is further supported by the fact that identical regions of the synthetase recognize both the operator and the tRNA anticodon arm. Based on these results, and recent structural data, we have constructed a computer-derived molecular model for the operator-threonyl-tRNA synthetase complex, which sheds light on several essential aspects of the regulatory mechanism.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/genetics , Threonine-tRNA Ligase/chemistry , Threonine-tRNA Ligase/metabolism , Binding Sites , Binding, Competitive , Evolution, Molecular , Gene Expression Regulation, Bacterial , Genes, Bacterial , Macromolecular Substances , Models, Molecular , Molecular Mimicry , Molecular Structure , Mutation , Operator Regions, Genetic , Protein Structure, Tertiary , Protein Subunits , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/metabolism , Threonine-tRNA Ligase/genetics
16.
J Biol Chem ; 278(4): 2723-30, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12435744

ABSTRACT

Human immunodeficiency virus (HIV) genomic RNA is packaged into virions as a dimer. The first step of dimerization is the formation of a kissing-loop complex at the so-called dimerization initiation site (DIS). We found an unexpected and fortuitous resemblance between the HIV-1 DIS kissing-loop complex and the eubacterial 16 S ribosomal aminoacyl-tRNA site (A site), which is the target of aminoglycoside antibiotics. Similarities exist not only at the primary and secondary structure level but also at the tertiary structure level, as revealed by comparison of the respective DIS and A site crystal structures. Gel shift, inhibition of lead-induced cleavage, and footprinting experiments showed that paromomycin and neomycin specifically bind to the kissing-loop complex formed by the DIS, with an affinity and a geometry similar to that observed for the A site. Modeling of the aminoglycoside-DIS complex allowed us to identify antibiotic modifications likely to increase the affinity and/or the specificity for the DIS. This could be a starting point for designing antiviral drugs against HIV-1 RNA dimerization.


Subject(s)
Anti-Bacterial Agents/pharmacology , HIV-1/metabolism , RNA, Viral , Ribosomes/metabolism , Binding Sites , Dimerization , Models, Molecular , Neomycin/pharmacology , Nucleic Acid Conformation , Paromomycin/pharmacology , Protein Binding , RNA/metabolism , Temperature , Ultraviolet Rays
17.
J Biol Chem ; 277(45): 43233-42, 2002 Nov 08.
Article in English | MEDLINE | ID: mdl-12194974

ABSTRACT

Initiation of human immunodeficiency virus type 1 (HIV-1) reverse transcription requires specific recognition between the viral RNA (vRNA), tRNA(3)(Lys), which acts as primer, and reverse transcriptase (RT). The specificity of this ternary complex is mediated by intricate interactions between the HIV-1 RNA and tRNA(3)(Lys). Here, we compared the relative importance of the secondary structure elements of this complex in the initiation process. To this aim, we used the previously published three-dimensional model of the initiation complex to rationally introduce a series of deletions and substitutions in the vRNA. When necessary, we used chemical probing to check the structure of the tRNA(3)(Lys)-mutant vRNA complexes. For each of them, we measured the binding affinity of RT and the kinetics of initial extension of tRNA(3)(Lys) and of synthesis of the (-) strand strong stop DNA. Our results were overall in keeping with the three-dimensional model of the initiation complex. Surprisingly, we found that disruption of the intermolecular template-primer interactions, which are not directly recognized by RT, more severely affected reverse transcription than deletions or disruption of one of the intramolecular helices to which RT directly binds. Perturbations of the highly constrained junction between the intermolecular helix formed by the primer binding site and the 3' end of tRNA(3)(Lys) and the helix immediately upstream also had dramatic effects on the initiation of reverse transcription. Taken together, our results demonstrate the overwhelming importance of the overall three-dimensional structure of the initiation complex and identify structural elements that constitute promising targets for anti-initiation-specific drugs.


Subject(s)
HIV Reverse Transcriptase/metabolism , HIV-1/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Base Sequence , DNA Primers , DNA Replication , Humans , Kinetics , Polymerase Chain Reaction , RNA, Transfer, Lys/genetics , RNA, Viral/metabolism , Transcription, Genetic
18.
J Mol Biol ; 320(5): 963-78, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12126618

ABSTRACT

Escherichia coli ribosomal protein S15 recognizes two RNA targets: a three-way junction in 16S rRNA and a pseudoknot structure on its own mRNA. Binding to mRNA occurs when S15 is expressed in excess over its rRNA target, resulting in an inhibition of translation start. The sole apparent similarity between the rRNA and mRNA targets is the presence of a G-U/G-C motif that contributes only modestly to rRNA binding but is essential for mRNA. To get more information on the structural determinants used by S15 to bind its mRNA target as compared to its rRNA site, we used site-directed mutagenesis, substitution by nucleotide analogs, footprinting experiments on both RNA and protein, and graphic modeling. The size of the mRNA-binding site could be reduced to 45 nucleotides, without loss of affinity. This short RNA preferentially folds into a pseudoknot, the formation of which depends on magnesium concentration and temperature. The size of the loop L2 that bridges the two stems of the pseudoknot through the minor groove could not be reduced below nine nucleotides. Then we showed that the pseudoknot recognizes the same side of S15 as 16S rRNA, although shielding a smaller surface area. It turned out that the G-U/G-C motif is recognized from the minor groove in both cases, and that the G-C pair is recognized in a very similar manner. However, the wobble G-U pair of the mRNA is not directly contacted by S15, as in rRNA, but is most likely involved in building a precise conformation of the RNA, essential for binding. Otherwise, unique specific features are utilized, such as the three-way junction in the case of 16S rRNA and the looped out A(-46) for the mRNA pseudoknot.


Subject(s)
RNA, Messenger/chemistry , RNA, Ribosomal/chemistry , Ribosomal Proteins/chemistry , Binding Sites , Cytosine , Escherichia coli , Guanosine , Models, Molecular , Nucleic Acid Conformation , Protein Structure, Tertiary , Uridine
19.
Nat Struct Biol ; 9(5): 343-7, 2002 May.
Article in English | MEDLINE | ID: mdl-11953757

ABSTRACT

Escherichia coli threonyl-tRNA synthetase (ThrRS) represses the translation of its own messenger RNA by binding to an operator located upstream of the initiation codon. The crystal structure of the complex between the core of ThrRS and the essential domain of the operator shows that the mRNA uses the recognition mode of the tRNA anticodon loop to initiate binding. The final positioning of the operator, upon which the control mechanism is based, relies on a characteristic RNA motif adapted to the enzyme surface. The finding of other thrS operators that have this conserved motif leads to a generalization of this regulatory mechanism to a subset of Gram-negative bacteria.


Subject(s)
Escherichia coli/enzymology , Protein Biosynthesis , RNA, Messenger/metabolism , Threonine-tRNA Ligase/chemistry , Threonine-tRNA Ligase/metabolism , Anticodon/genetics , Base Sequence , Crystallography, X-Ray , Escherichia coli/genetics , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Conformation , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Sequence Alignment , Structure-Activity Relationship
20.
J Biol Chem ; 277(21): 18611-8, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11901149

ABSTRACT

Azidothymidine (AZT) is a widely used inhibitor of type 1 human immunodeficiency virus reverse transcriptase (RT) that acts as chain terminator. Upon treatment, mutations conferring AZT resistance to RT are gradually selected. It has been shown that resistant RT is able to unblock the AZT-terminated primer by an ATP-dependent mechanism. However, this resistance mechanism has only been demonstrated for DNA-dependent DNA elongation. Here, we compared the AZT resistance of mutant RT during DNA elongation on DNA and RNA templates. We showed that, during DNA elongation, primer unblocking and rescue of DNA synthesis take place with similar rate constants on DNA and RNA templates. However, the fraction of a primer eventually repaired during RNA-dependent DNA synthesis is 2x lower compared with that of DNA-dependent synthesis, leading to reduced resistance. We also compared the initiation of reverse transcription, which uses tRNA(3)(Lys) as a primer and displays characteristic kinetic features, and the subsequent RNA-dependent elongation. Unlike during elongation, resistant RT was unable to unblock the AZT-terminated primer during initiation of (-) DNA strand synthesis. Our results demonstrate that the efficiency of primer unblocking conferred by the AZT resistance mutations greatly vary during the different steps of the provirus synthesis. These results also suggest that inhibitors specifically targeting the initiation of reverse transcription might prove to be advantageous, as compared with elongation inhibitors.


Subject(s)
DNA Replication , HIV Reverse Transcriptase/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Zidovudine/pharmacology , Base Sequence , DNA Primers , Drug Resistance, Microbial , Kinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...