Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Chem Biol ; 22(6): 712-23, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26051216

ABSTRACT

The cellular DNA repair hRAD51 protein has been shown to restrict HIV-1 integration both in vitro and in vivo. To investigate its regulatory functions, we performed a pharmacological analysis of the retroviral integration modulation by hRAD51. We found that, in vitro, chemical activation of hRAD51 stimulates its integration inhibitory properties, whereas inhibition of hRAD51 decreases the integration restriction, indicating that the modulation of HIV-1 integration depends on the hRAD51 recombinase activity. Cellular analyses demonstrated that cells exhibiting high hRAD51 levels prior to de novo infection are more resistant to integration. On the other hand, when hRAD51 was activated during integration, cells were more permissive. Altogether, these data establish the functional link between hRAD51 activity and HIV-1 integration. Our results highlight the multiple and opposite effects of the recombinase during integration and provide new insights into the cellular regulation of HIV-1 replication.


Subject(s)
HIV-1/physiology , Rad51 Recombinase/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/chemistry , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , DNA/chemistry , DNA/metabolism , DNA Repair , Gene Expression/drug effects , HEK293 Cells , Humans , Morpholines/chemistry , Morpholines/metabolism , Morpholines/pharmacology , Protein Binding , Pyrroles/chemistry , Pyrroles/metabolism , Pyrroles/pharmacology , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , Stilbenes/chemistry , Stilbenes/metabolism , Stilbenes/pharmacology , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology , Virus Internalization/drug effects , Virus Replication/drug effects
2.
Nucleic Acids Res ; 42(20): 12352-66, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25332402

ABSTRACT

G-rich nucleic acids can form non-canonical G-quadruplex structures (G4s) in which four guanines fold in a planar arrangement through Hoogsteen hydrogen bonds. Although many biochemical and structural studies have focused on DNA sequences containing successive, adjacent guanines that spontaneously fold into G4s, evidence for their in vivo relevance has recently begun to accumulate. Complete sequencing of the human genome highlighted the presence of ∼300,000 sequences that can potentially form G4s. Likewise, the presence of putative G4-sequences has been reported in various viruses genomes [e.g., Human immunodeficiency virus (HIV-1), Epstein-Barr virus (EBV), papillomavirus (HPV)]. Many studies have focused on telomeric G4s and how their dynamics are regulated to enable telomere synthesis. Moreover, a role for G4s has been proposed in cellular and viral replication, recombination and gene expression control. In parallel, DNA aptamers that form G4s have been described as inhibitors and diagnostic tools to detect viruses [e.g., hepatitis A virus (HAV), EBV, cauliflower mosaic virus (CaMV), severe acute respiratory syndrome virus (SARS), simian virus 40 (SV40)]. Here, special emphasis will be given to the possible role of these structures in a virus life cycle as well as the use of G4-forming oligonucleotides as potential antiviral agents and innovative tools.


Subject(s)
Antiviral Agents/chemistry , DNA, Viral/chemistry , G-Quadruplexes , RNA, Viral/chemistry , Genome, Human , Humans
3.
Biochimie ; 95(4): 759-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23160073

ABSTRACT

DNA replication occurs in various compartments of eukaryotic cells such as the nuclei, mitochondria and chloroplasts, the latter of which is used in plants and algae. Replication appears to be simpler in the mitochondria than in the nucleus where multiple DNA polymerases, which are key enzymes for DNA synthesis, have been characterized. In mammals, only one mitochondrial DNA polymerase (pol γ) has been described to date. However, in the mitochondria of the yeast Saccharomyces cerevisiae, we have found and characterized a second DNA polymerase. To identify this enzyme, several biochemical approaches such as proteinase K treatment of sucrose gradient purified mitochondria, analysis of mitoplasts, electron microscopy and the use of mitochondrial and cytoplasmic markers for immunoblotting demonstrated that this second DNA polymerase is neither a nuclear or cytoplasmic contaminant nor a proteolytic product of pol γ. An improved purification procedure and the use of mass spectrometry allowed us to identify this enzyme as DNA polymerase α. Moreover, tagging DNA polymerase α with a fluorescent probe demonstrated that this enzyme is localized both in the nucleus and in the organelles of intact yeast cells. The presence of two replicative DNA polymerases may shed new light on the mtDNA replication process in S. cerevisiae.


Subject(s)
DNA Polymerase I/chemistry , DNA Polymerase I/metabolism , Mitochondria/enzymology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , DNA Polymerase I/genetics , DNA Replication , DNA, Mitochondrial/biosynthesis , Endopeptidase K/metabolism , Mitochondria/metabolism , Mutation , Protein Transport , Saccharomyces cerevisiae/genetics
4.
J Biomed Biotechnol ; 2012: 549020, 2012.
Article in English | MEDLINE | ID: mdl-22778552

ABSTRACT

Despite being simple eukaryotic organisms, the yeasts Saccharomyces cerevisiae and Schizosaccharomyces pombe have been widely used as a model to study human pathologies and the replication of human, animal, and plant viruses, as well as the function of individual viral proteins. The complete genome of S. cerevisiae was the first of eukaryotic origin to be sequenced and contains about 6,000 genes. More than 75% of the genes have an assigned function, while more than 40% share conserved sequences with known or predicted human genes. This strong homology has allowed the function of human orthologs to be unveiled starting from the data obtained in yeast. RNA plant viruses were the first to be studied in yeast. In this paper, we focus on the use of the yeast model to study the function of the proteins of human immunodeficiency virus type 1 (HIV-1) and the search for its cellular partners. This human retrovirus is the cause of AIDS. The WHO estimates that there are 33.4 million people worldwide living with HIV/AIDS, with 2.7 million new HIV infections per year and 2.0 million annual deaths due to AIDS. Current therapy is able to control the disease but there is no permanent cure or a vaccine. By using yeast, it is possible to dissect the function of some HIV-1 proteins and discover new cellular factors common to this simple cell and humans that may become potential therapeutic targets, leading to a long-lasting treatment for AIDS.


Subject(s)
HIV-1/physiology , Yeasts/physiology , Animals , Fungal Proteins/genetics , Fungal Proteins/physiology , HIV-1/genetics , Humans , Viral Proteins/genetics , Viral Proteins/physiology , Yeasts/genetics
5.
PLoS One ; 2(12): e1315, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-18091989

ABSTRACT

IntI1 integrase is a tyrosine recombinase involved in the mobility of antibiotic resistance gene cassettes within bacterial class 1 integrons. Recent data have shown that its recombination specifically involves the bottom strand of the attC site, but the exact mechanism of the reaction is still unclear. An efficient in vitro assay is still required to better characterize the biochemical properties of the enzyme. In this report we describe for the first time an in vitro system partially reproducing the activity of a recombinant pure IntI1. This new assay, which constitutes the only available in vitro model of recombination by IntI1, was used to determine whether this enzyme might be the sole bacterial protein required for the recombination process. Results show that IntI1 possesses all the features needed for performing recombination between attI and attC sites. However, differences in the in vitro intermolecular recombination efficiencies were found according to the target sites and were correlated with DNA affinities of the enzyme but not with in vivo data. The differential affinity of the enzyme for each site, its capacity to bind to a single-stranded structure at the attC site and the recombination observed with single-stranded substrates unambiguously confirm that it constitutes an important intermediary in the reaction. Our data strongly suggest that the enzyme possesses all the functions for generating and/or recognizing this structure even in the absence of other cellular factors. Furthermore, the in vitro assay reported here constitutes a powerful tool for the analysis of the recombination steps catalyzed by IntI1, its structure-function studies and the search for specific inhibitors.


Subject(s)
Integrases/metabolism , Pseudomonas aeruginosa/enzymology , Vibrio cholerae/enzymology , Base Sequence , Catalysis , DNA Primers , Integrases/isolation & purification , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombination, Genetic
6.
Oligonucleotides ; 17(2): 151-65, 2007.
Article in English | MEDLINE | ID: mdl-17638520

ABSTRACT

We have previously described how a 16 nucleotides ODN (termed 93del) is capable of inhibiting the activity of recombinant integrase in a cell-free system as well as HIV-1 replication in human-infected cells with IC(50) in the low nanomolar range. Intracellular HIV-1 replication was inhibited when the ODN was added at the onset of infection. These results raise several questions. Is a naked ODN able to enter the cell? Does the virus play a role in ODN entry? The uptake of several ODNs (93del, 60del(sc), TBA, T30923) was evaluated and then tracked by labeling the ODN with a fluorescent dye and assessing its intracellular localization by confocal microscopy. A significant level of cellular uptake of free ODN was observed in several cell lines: HeLa epithelial cells, Huh7 hepatic cells, and H9 lymphocytes, and was detected for all ODNs tested except for TBA. Striking differences were observed when naked ODNs were added to cell in the presence or absence of the virus. When HIV-1 virions were present a sharp increase in cellular fluorescence was observed. These results strongly suggest a role for HIV-1 virions in the uptake of certain ODNs.


Subject(s)
HIV-1/physiology , Oligonucleotides/metabolism , Cell Line , Flow Cytometry , Fluorescent Dyes , HIV Integrase/metabolism , HIV Integrase Inhibitors/pharmacology , HIV-1/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Oligonucleotides/pharmacology , Virion/physiology
7.
Nucleic Acids Res ; 35(3): 975-87, 2007.
Article in English | MEDLINE | ID: mdl-17259219

ABSTRACT

HIV-1 integrase (IN) catalyses integration of a DNA copy of the viral genome into the host genome. Specific interactions between retroviral IN and long terminal repeats (LTR) are required for this insertion. To characterize quantitatively the influence of the determinants of DNA substrate specificity on the oligomerization status of IN, we used the small-angle X-ray scattering (SAXS) technique. Under certain conditions in the absence of ODNs IN existed only as monomers. IN preincubation with specific ODNs led mainly to formation of dimers, the relative amount of which correlated well with the increase in the enzyme activity in the 3'-processing reaction. Under these conditions, tetramers were scarce. Non-specific ODNs stimulated formation of catalytically inactive dimers and tetramers. Complexes of monomeric, dimeric and tetrameric forms of IN with specific and non-specific ODNs had varying radii of gyration (R(g)), suggesting that the specific sequence-dependent formation of IN tetramers can probably occur by dimerization of two dimers of different structure. From our data we can conclude that the DNA-induced oligomerization of HIV-1 IN is probably of importance to provide substrate specificity and to increase the enzyme activity.


Subject(s)
DNA/chemistry , HIV Integrase/chemistry , HIV-1/enzymology , Nucleoproteins/chemistry , DNA/metabolism , Dimerization , HIV Integrase/metabolism , Kinetics , Nucleoproteins/metabolism , Oligodeoxyribonucleotides/chemistry , Scattering, Small Angle , Substrate Specificity , X-Ray Diffraction
8.
Biochimie ; 87(9-10): 911-9, 2005.
Article in English | MEDLINE | ID: mdl-16164998

ABSTRACT

Several in vitro strategies have been developed to selectively screen for nucleic acid sequences that bind to specific proteins. We previously used the SELEX procedure to search for aptamers against HIV-1 RNase H activity associated with reverse transcriptase (RT) and human RNase H1. Aptamers containing G-rich sequences were selected in both cases. To investigate whether the interaction with G-rich oligonucleotides (ODNs) was a characteristic of these enzymes, a second in vitro selection was performed with an isolated RNase H domain of HIV-1 RT (p15) as a target and a new DNA library. In this work we found that the second SELEX led again to the isolation of G-rich aptamers. But in contrast to the first selection, these latter ODNs were not able to inhibit the RNase H activity of either the p15 domain or the RNase H embedded in the complete RT. On the other hand, the aptamers from the first SELEX that were inhibitors of the RT-associated RNase H did not inhibit the activity of the isolated p15 domain. This suggests that the active conformation of both RNase H domains is different according to the presence or absence of the DNA polymerase domain. HIV-1 RNase H and integrase both belong to the phosphotransferase family and share structural similarities. An interesting result was obtained when the DNA aptamers initially raised against p15 RNase H were assayed against HIV-1 integrase. In contrast to RNase H, the HIV-1 integrase was inhibited by these aptamers. Our results point out that prototype structures can be exploited to develop inhibitors of two related enzymes.


Subject(s)
Aptamers, Nucleotide/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/drug effects , HIV-1/enzymology , Oligonucleotides/metabolism , Ribonuclease H/drug effects , Aptamers, Nucleotide/chemistry , Base Sequence , HIV Integrase/chemistry , HIV Integrase/isolation & purification , HIV Long Terminal Repeat , Humans , Oligonucleotides/chemistry , Ribonuclease H/chemistry , Ribonuclease H/metabolism , Templates, Genetic
9.
FEBS J ; 272(15): 3872-86, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16045758

ABSTRACT

In our attempt to obtain further information on the replication mechanism of the hepatitis C virus (HCV), we have studied the role of sequences at the 3'-end of HCV minus-strand RNA in the initiation of synthesis of the viral genome by viral RNA-dependent RNA polymerase (RdRp). In this report, we investigated the template and binding properties of mutated and deleted RNA fragments of the 3'-end of the minus-strand HCV RNA in the presence of viral polymerase. These mutants were designed following the newly established secondary structure of this viral RNA fragment. We showed that deletion of the 3'-SL-A1 stem loop significantly reduced the level of RNA synthesis whereas modifications performed in the SL-B1 stem loop increased RNA synthesis. Study of the region encompassing the 341 nucleotides of the 3'-end of the minus-strand RNA shows that these two hairpins play a very limited role in binding to the viral polymerase. On the contrary, deletions of sequences in the 5'-end of this fragment greatly impaired both RNA synthesis and RNA binding. Our results strongly suggest that several domains of the 341 nucleotide region of the minus-strand 3'-end interact with HCV RdRp during in vitro RNA synthesis, in particular the region located between nucleotides 219 and 239.


Subject(s)
Hepacivirus/enzymology , Hepacivirus/genetics , RNA, Antisense/biosynthesis , RNA, Viral/biosynthesis , Templates, Genetic , Viral Nonstructural Proteins/metabolism , 3' Untranslated Regions/metabolism , Base Sequence , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Hepacivirus/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Protein Binding/genetics , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Sequence Analysis, RNA , Sequence Deletion
10.
J Ethnopharmacol ; 99(1): 83-91, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15848024

ABSTRACT

Seventeen aqueous and methanol extracts from nine South African medicinal plants, ethnobotanically selected, were screened for inhibitory properties against HIV-1 reverse transcriptase (RT). Isolated compounds were additionally evaluated on HIV-1 integrase (IN). The strongest inhibition against the RNA-dependent-DNA polymerase (RDDP) activity of RT was observed with the methanol extract of the stem-bark of Peltophorum africanum Sond. (Fabaceae) (IC(50) 3.5 microg/ml), while the methanol extract of the roots of Combretum molle R.Br. ex G. Don (Combretaceae) was the most inhibitory on the ribonuclease H (RNase H) activity (IC(50) 9.7 microg/ml). The known compounds bergenin and catechin, and a red coloured gallotannin composed of meta-depside chains of gallic and protocatechuic acids esterified to a 1-O-isobutyroly-beta-D-glucopyranose core, were isolated from the methanol extract of the roots and stem-bark of Peltophorum africanum. The gallotannin inhibited the RDDP and RNase H functions of RT with IC(50) values of 6.0 and 5.0 microM, respectively, and abolished the 3'-end processing activity of IN at 100 microM. Catechin showed no effect on RT but had a moderate activity on HIV-1 IN. Bergenin was inactive on both enzymes. The aqueous and methanol extracts were non-toxic in a HeLaP4 cell line at a concentration of 400 microg/ml.


Subject(s)
Anti-HIV Agents/isolation & purification , Anti-HIV Agents/pharmacology , HIV Integrase Inhibitors/isolation & purification , HIV Integrase Inhibitors/pharmacology , HIV Integrase , HIV Reverse Transcriptase , Plants, Medicinal/chemistry , Reverse Transcriptase Inhibitors/isolation & purification , Reverse Transcriptase Inhibitors/pharmacology , Cell Survival/drug effects , Combretum/chemistry , DNA, Viral/drug effects , Ethanol , Humans , Medicine, African Traditional , Plant Extracts/chemistry , Plant Extracts/pharmacology , Solvents , South Africa , Tumor Cells, Cultured , Water
11.
Nucleic Acids Res ; 33(3): 977-86, 2005.
Article in English | MEDLINE | ID: mdl-15718297

ABSTRACT

The oligomeric state of active human immunodeficiency virus type 1 (HIV-1) integrase (IN) has not been clearly elucidated. We analyzed the activity of the different purified oligomeric forms of recombinant IN obtained after stabilization by platinum crosslinking. The crosslinked tetramer isolated by gel chromatography was able to catalyze the full-site integration of the two viral LTR ends into a target DNA in vitro, whereas the isolated dimeric form of the enzyme was involved in the processing and integration of only one viral end. Accurate concerted integration by IN tetramers was confirmed by cloning and sequencing. Kinetic studies of DNA-integrase complexes led us to propose a model explaining the formation of an active complex. Our data suggest that the tetrameric IN bound to the viral DNA ends is the minimal complex involved in the concerted integration of both LTRs and should be the oligomeric form targeted by future inhibitors.


Subject(s)
HIV Integrase/metabolism , HIV-1/enzymology , Cross-Linking Reagents , DNA/metabolism , HIV Integrase/genetics , HIV Integrase/isolation & purification , HIV Long Terminal Repeat , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Yeasts/genetics
12.
Biochemistry ; 43(41): 13097-105, 2004 Oct 19.
Article in English | MEDLINE | ID: mdl-15476403

ABSTRACT

Human immunodeficiency virus type 1 integrase catalyzes the integration of proviral DNA into the infected cell genome, so it is an important potential target for antiviral drug design. In an attempt to search for peptides that specifically interact with integrase (IN) and inhibit its function, we used an in vitro selection procedure, the phage display technique. A phage display library of random heptapeptides was used to screen for potential peptide ligands of HIV-1 IN. Several phage clones were identified that specifically bound IN. Two of the selected peptides (FHNHGKQ and HLEHLLF) exhibited a high affinity for IN and were chemically synthesized. High affinity was confirmed by a displacement assay which showed that these two synthetic peptides were able to compete with the phages expressing the corresponding peptide. These agents were assayed on the in vitro IN activities. While none of them inhibited the 3'-processing reaction, the FHNHGKQ peptide was found to be an inhibitor of the strand transfer reaction. Despite its high affinity for IN, the HLEHLLF peptide selected and assayed under the same conditions was unable to inhibit this reaction. We showed that the FHNHGKQ peptide inhibits specifically the strand transfer activity by competing with the target DNA for binding to IN. These IN-binding agents could be used as a base for developing new anti-integrase compounds as well as for structural studies of the still unknown three-dimensional structure of the entire integrase molecule.


Subject(s)
Bacteriophage M13 , HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , Oligopeptides/chemistry , Peptide Library , Transcription, Genetic , Virus Integration , Binding, Competitive , Capsid Proteins , Catalysis , Catalytic Domain , DNA-Binding Proteins/chemical synthesis , DNA-Binding Proteins/metabolism , Dimerization , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Protein Binding , Substrate Specificity , Viral Fusion Proteins/chemical synthesis , Viral Fusion Proteins/metabolism
13.
Gene ; 322: 157-68, 2003 Dec 11.
Article in English | MEDLINE | ID: mdl-14644507

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) integrase (IN) catalyzes the insertion of the viral genome into the host cell DNA, an essential reaction during the retroviral cycle. We described previously that expression of HIV-1 IN in some yeast strains may lead to the emergence of a lethal phenotype which was not observed when the catalytically crucial residues D, D, (35)E were mutated. The lethal effect in yeast seems to be related to the mutagenic effect of the recombinant HIV-1 IN, most probably via the non-sequence-specific endonucleolytic activity carried by this enzyme. This non-sequence-specific endonuclease activity was further characterized. Although the enzyme was active on DNA substrates devoid of viral long terminal repeat (LTR) sequences, the presence of LTR regions stimulated significantly this activity. Genetic experiments were designed to show that both the mutagenic effect and the level of recombination events were affected in cells expressing the active retroviral enzyme, while expression of the mutated inactive IN D116A has no significant effect. A close interaction was demonstrated between integrase activity and in vivo/in vitro recombination process, suggesting that retroviral integration and recombination mechanism are linked in the infected cell. Our results show that the yeast system is a powerful cellular model to study the non-sequence-specific endonucleolytic activity of IN. Its characterization is essential since this activity might represent a very important step in the retroviral infectious cycle and would provide further insights into the function of IN. Indeed, effectors of this activity should be sought as potential antiviral agents since stimulation of this enzymatic activity would induce the destruction of early synthesized proviral DNA.


Subject(s)
DNA Repair , HIV Integrase/metabolism , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Cell Division/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Deoxyribonuclease I/metabolism , Diploidy , Gene Expression Regulation, Enzymologic , HIV Integrase/genetics , HIV Long Terminal Repeat/genetics , Haploidy , Mutation , Phenotype , Plasmids/genetics , Saccharomyces cerevisiae/growth & development
14.
Virology ; 314(1): 206-20, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14517074

ABSTRACT

We describe oligonucleotides (ODNs) that inhibit hepatitis C virus (HCV) RNA synthesis in vitro. From a series of 13 ODNs complementary to the 3'-end of the minus-strand HCV RNA, only 4 inhibited RNA synthesis with IC(50) values lower than 1 microM. The inhibition was sequence-specific, since no effect was observed when the ODNs were used with a noncomplementary template. The introduction of a 2'-O-methyl modification increased the inhibitor activity 11-fold (IC(50) = 50 nM) in just 1 (ODN7) of the 4 inhibitory ODNs. ODNs did not inhibit RNA synthesis by interfering with the elongation process as no short RNAs products were detected. We also show that ODN7 did not prevent binding of NS5B to the template or cause polymerase trapping by the duplex RNA/ODN. Our data demonstrate that ODN7 inhibits the initiation process, most probably by modifying structural features present at the 3'-end of the minus-strand RNA.


Subject(s)
3' Untranslated Regions/chemistry , Hepacivirus/genetics , Oligonucleotides/pharmacology , RNA, Viral/biosynthesis , RNA, Viral/drug effects , RNA-Dependent RNA Polymerase/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Genetic Complementation Test , Molecular Sequence Data , Oligonucleotides/chemistry , RNA, Viral/chemistry , RNA, Viral/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
15.
Biochemistry ; 42(30): 9235-47, 2003 Aug 05.
Article in English | MEDLINE | ID: mdl-12885259

ABSTRACT

Specific interactions between retroviral integrase (IN) and long terminal repeats are required for insertion of viral DNA into the host genome. To characterize quantitatively the determinants of substrate specificity, we used a method based on a stepwise increase in ligand complexity. This allowed an estimation of the relative contributions of each nucleotide from oligonucleotides to the total affinity for IN. The interaction of HIV-1 integrase with specific (containing sequences from the LTR) or nonspecific oligonucleotides was analyzed using a thermodynamic model. Integrase interacted with oligonucleotides through a superposition of weak contacts with their bases, and more importantly, with the internucleotide phosphate groups. All these structural components contributed in a combined way to the free energy of binding with the major contribution made by the conserved 3'-terminal GT, and after its removal, by the CA dinucleotide. In contrast to nonspecific oligonucleotides that inhibited the reaction catalyzed by IN, specific oligonucleotides enhanced the activity, probably owing to the effect of sequence-specific ligands on the dynamic equilibrium between the oligomeric forms of IN. However, after preactivation of IN by incubation with Mn(2+), the specific oligonucleotides were also able to inhibit the processing reaction. We found that nonspecific interactions of IN with DNA provide approximately 8 orders of magnitude in the affinity (Delta G degrees approximately equal to -10.3 kcal/mol), while the relative contribution of specific nucleotides of the substrate corresponds to approximately 1.5 orders of magnitude (Delta G degrees approximately equal to - 2.0 kcal/mol). Formation of the Michaelis complex between IN and specific DNA cannot by itself account for the major contribution of enzyme specificity, which lies in the k(cat) term; the rate is increased by more than 5 orders of magnitude upon transition from nonspecific to specific oligonucleotides.


Subject(s)
DNA, Viral/chemistry , DNA, Viral/metabolism , HIV Integrase/chemistry , HIV Integrase/genetics , HIV-1/enzymology , HIV-1/genetics , Thermodynamics , Transformation, Genetic , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Enzyme Activation , Humans , Kinetics , Models, Chemical , Nucleic Acid Heteroduplexes/genetics , Nucleic Acid Heteroduplexes/metabolism , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Protein Binding , Substrate Specificity
16.
Nucleic Acids Res ; 31(4): 1275-81, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12582247

ABSTRACT

The hepatitis C virus (HCV) 5' untranslated region (UTR) has been extensively studied with regard to its internal ribosomal entry site (IRES) activity. In this work we present results suggesting the existence of a strong promoter activity carried by the DNA sequence corresponding to the HCV 5' UTR. This activity was not detected when the HCV 5' UTR sequence was replaced by HCV 3' UTR or poliovirus 5' UTR sequences. These results were further confirmed by using bicistronic constructions. We demonstrated the presence of an mRNA initiated in this 5' UTR sequence and located the initiation site by the 5' RACE method at nucleotide 67. Furthermore, northern experiments and flow cytometry analysis showed the unambiguous activity of such a promoter sequence in stably transfected cells. Our results strongly suggest that the data obtained using bicistronic DNA constructs carrying the HCV 5' UTR should be analyzed not only at the translational but also at the transcriptional level.


Subject(s)
5' Untranslated Regions/genetics , Hepacivirus/genetics , Promoter Regions, Genetic/genetics , 5' Untranslated Regions/chemistry , Base Sequence , DNA, Complementary/genetics , Gene Expression , Genome, Viral , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , Transcription Initiation Site , Transcription, Genetic , Transfection , Tumor Cells, Cultured
17.
Oligonucleotides ; 13(6): 455-63, 2003.
Article in English | MEDLINE | ID: mdl-15025912

ABSTRACT

The RNA-dependent RNA polymerase (NS5B) of the hepatitis C virus (HCV) plays a key role in the life cycle of the virus. In order to find inhibitors of the HCV polymerase, we screened a library of 81 nucleotide (nt)-long synthetic DNA containing 35 random nucleotides by the Systematic Evolution of Ligands by Exponential enrichment (SELEX) approach. Thirty ligands selected for their binding affinity to the NS5B were classified into four groups on the basis of their sequence homologies. Among the selected molecules, two were able to inhibit in vitro the polymerase activity of the HCV NS5B. These aptamers appeared to be specific for HCV polymerase, as no inhibition of poliovirus 3D polymerase activity was observed. The binding and inhibitory potential of one aptamer (27v) was associated with the 35 nt-long variable region. This oligonucleotide displayed an apparent dissociation constant (K(d)) in the nanomolar range. Our results showed that it was able to compete with RNA templates corresponding to the 3'-ends of the (+) and the (-) HCV RNA for binding to the polymerase. The fact that a DNA aptamer could interfere with the binding of natural templates of the enzyme could help in performing structure-function analysis of the NS5B and might constitute a basis for further structure-based drug design of this crucial enzyme of HCV replication.


Subject(s)
Hepacivirus/genetics , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/pharmacology , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Base Sequence , Electrophoretic Mobility Shift Assay , Gene Library , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , RNA, Messenger/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Transcription, Genetic/drug effects , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
18.
J Mol Biol ; 318(1): 45-58, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-12054767

ABSTRACT

The retroviral encoded protein integrase (IN) is required for the insertion of the human immunodeficiency virus type 1 (HIV-1) proviral DNA into the host genome. In spite of the crucial role played by IN in the retroviral life cycle, which makes this enzyme an attractive target for the development of new anti-AIDS agents, very few inhibitors have been described and none seems to have a potential use in anti-HIV therapy. To obtain potent and specific IN inhibitors, we used the two-hybrid system to isolate short peptides. Using HIV-1 IN as a bait and a yeast genomic library as the source of inhibitory peptides (prey), we isolated a 33-mer peptide (I33) that bound tightly to the enzyme. I33 inhibited both in vitro IN activities, i.e. 3' end processing and strand transfer. Further analysis led us to select a shorter peptide, EBR28, corresponding to the N-terminal region of I33. Truncated variants showed that EBR28 interacted with the catalytic domain of IN interfering with the binding of the DNA substrate. Alanine single substitution of each EBR28 residue (alanine scanning) allowed the identification of essential amino acids involved in the inhibition. The EBR28 NMR structure shows that this peptide adopts an alpha-helical conformation with amphipathic properties. Additionally, EBR28 showed a significant antiviral effect when assayed on HIV-1 infected human cells. Thus, this potentially important short lead peptide may not only be helpful to design new anti-HIV agents, but also could prove very useful in further studies of the structural and functional characteristics of HIV-1 IN.


Subject(s)
HIV Integrase Inhibitors/chemistry , HIV-1/enzymology , Oligopeptides/chemistry , Peptides/chemistry , Alanine/metabolism , Algorithms , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Catalysis , Catalytic Domain , DNA, Viral/chemistry , DNA, Viral/genetics , DNA, Viral/metabolism , HIV Integrase Inhibitors/pharmacology , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Oligopeptides/pharmacology , Peptides/isolation & purification , Peptides/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Tumor Cells, Cultured , Yeasts/genetics
19.
Curr Pharm Des ; 8(8): 595-614, 2002.
Article in English | MEDLINE | ID: mdl-11945161

ABSTRACT

The rapid spread of the AIDS epidemic has stimulated the search for new agents able to arrest the replication of the causative virus, HIV. The best strategy for AIDS treatment involves a combination therapy using inhibitors of reverse transcriptase and protease. However, the emergence of HIV-1 strains resistant to these drugs and their cytotoxicity requires the synthesis and the biochemical and cellular characterization of new antiviral drugs, as well as the development of newer strategies and viral targets. In addition to reverse transcriptase and protease, other retroviral enzymes acting in the replicative cycle of HIV-1 are potential targets for chemotherapeutic intervention. Like all retroviruses, HIV-1 requires the integration of the proviral double-stranded DNA, arising from the reverse transcription step, into the host chromosome for its efficient replication, maintenance of a stably infected state and productive infection. DNA integration is carried out by integrase so this enzyme represents a key area in developing new anti-retroviral therapy. Another novel enzymatic target concerns the RNase H activity associated with the retroviral reverse transcriptase, since a functional RNase H is essential for retroviral replication. Inhibitors against HIV-1 integrase and RNase H having potential therapeutical propeties have not yet been described. We focus this review on the properties of inhibitors of reverse transcriptase and integrase. Some of these antiviral agents have been known for several years while others are emerging as new promising strategies based on the use of oligonucleotides with special emphasis on the SELEX approach, peptides and retrovirucides.


Subject(s)
Drug Design , HIV Integrase Inhibitors/chemistry , HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/chemistry , Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/chemistry , Anti-HIV Agents/therapeutic use , Drug Evaluation, Preclinical , Drug Resistance, Viral , Drug Therapy, Combination , HIV Integrase/metabolism , HIV Integrase Inhibitors/therapeutic use , Humans , Molecular Structure , Oligonucleotides/chemistry , Oligonucleotides/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Ribonuclease H/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...