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











Publication year range
1.
Bioorg Med Chem ; 27(16): 3595-3604, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31285097

ABSTRACT

Inspired by bioactive biaryl-containing natural products found in plants and the marine environment, a series of synthetic compounds belonging to the azaBINOL chiral ligand family was evaluated for antiviral activity against HIV-1. Testing of 39 unique azaBINOLs and two BINOLs in a single-round infectivity assay resulted in the identification of three promising antiviral compounds, including 7-isopropoxy-8-(naphth-1-yl)quinoline (azaBINOL B#24), which exhibited low-micromolar activity without associated cytotoxicity. The active compounds and several close structural analogues were further tested against three different HIV-1 envelope pseudotyped viruses as well as in a full-virus replication system (EASY-HIT). The in vitro studies indicated that azaBINOL B#24 acts on early stages of viral replication before viral assembly and budding. Next we explored B#24's activity against HIV-1 reverse transcriptase (RT) and individually tested for polymerase and RNase H activity. The azaBINOL B#24 inhibits RNase H activity and binds directly to the HIV-1 RT enzyme. Additionally, we observe additive inhibitory activity against pseudotyped viruses when B#24 is dosed in competition with the clinically used non-nucleoside reverse transcriptase inhibitor (NNRTI) efavirenz. When tested against a multi-drug resistant HIV-1 isolate with drug resistance associated mutations in regions encoding for HIV-1 RT and protease, B#24 only exhibits a 5.1-fold net decrease in IC50 value, while efavirenz' activity decreases by 7.6-fold. These results indicate that azaBINOL B#24 is a potentially viable, novel lead for the development of new HIV-1 RNase H inhibitors. Furthermore, this study demonstrates that the survey of libraries of synthetic compounds, designed purely with the goal of facilitating chemical synthesis in mind, may yield unexpected and selective drug leads for the development of new antiviral agents.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV-1/drug effects , Quinolines/therapeutic use , Ribonuclease H/drug effects , Anti-HIV Agents/pharmacology , Humans , Quinolines/pharmacology
2.
Antiviral Res ; 143: 205-217, 2017 07.
Article in English | MEDLINE | ID: mdl-28450058

ABSTRACT

We previously reported low sensitivity of the hepatitis B virus (HBV) ribonuclease H (RNaseH) enzyme to inhibition by N-hydroxyisoquinolinedione (HID) compounds. Subsequently, our biochemical RNaseH assay was found to have a high false negative rate for predicting HBV replication inhibition, leading to underestimation of the number of HIDs that inhibit HBV replication. Here, 39 HID compounds and structurally related polyoxygenated heterocycles (POH), N-hydroxypyridinediones (HPD), and flutimides were screened for inhibition of HBV replication in vitro. Inhibiting the HBV RNaseH preferentially blocks synthesis of the positive-polarity DNA strand and causes accumulation of RNA:DNA heteroduplexes. Eleven HIDs and one HPD preferentially inhibited HBV positive-polarity DNA strand accumulation. EC50s ranged from 0.69 µM to 19 µM with therapeutic indices from 2.4 to 71. Neither the HIDs nor the HPD had an effect on the ability of the polymerase to elongate DNA strands in capsids. HBV RNaseH inhibition by the HIDs was confirmed with an improved RNaseH assay and by detecting accumulation RNA:DNA heteroduplexes in HBV capsids from cells treated with a representative HID. Therefore, the HID scaffold is more promising for anti-HBV drug discovery than we originally reported, and the HPD scaffold may hold potential for antiviral development. The preliminary structure-activity relationship will guide optimization of the HID/HPDs as HBV inhibitors.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Antiviral Agents/chemistry , Hepatitis B virus/drug effects , Virus Replication/drug effects , Animals , Antiviral Agents/administration & dosage , Capsid Proteins/genetics , Cell Line, Tumor , Chlorocebus aethiops , DNA Replication/drug effects , DNA, Viral/drug effects , Drug Discovery , Drug Evaluation, Preclinical , Hepatitis B/virology , Hepatitis B virus/enzymology , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , Microbial Sensitivity Tests , Piperazines/pharmacology , Ribonuclease H/drug effects , Structure-Activity Relationship , Vero Cells
3.
J Struct Biol ; 187(2): 119-128, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24972277

ABSTRACT

RNase H1 from extreme halophilic archaeon Halobacterium sp. NRC-1 (Halo-RNase H1) requires ⩾2M NaCl, ⩾10mM MnCl2, or ⩾300mM MgCl2 for folding. To understand the structural basis for this salt-dependent folding of Halo-RNase H1, the crystal structure of Halo-RNase H1 was determined in the presence of 10mM MnCl2. The structure of Halo-RNase H1 highly resembles those of metagenome-derived LC11-RNase H1 and Sulfolobus tokodaii RNase H1 (Sto-RNase H1), except that it contains two Mn(2+) ions at the active site and has three bi-aspartate sites on its surface. To examine whether negative charge repulsion at these sites are responsible for low-salt denaturation of Halo-RNase H1, a series of the mutant proteins of Halo-RNase H1 at these sites were constructed. The far-UV CD spectra of these mutant proteins measured in the presence of various concentrations of NaCl suggest that these mutant proteins exist in an equilibrium between a partially folded state and a folded state. However, the fraction of the protein in a folded state is nearly 0% for the active site mutant, 40% for the bi-aspartate site mutant, and 70% for the mutant at both sites in the absence of salt. The active site mutant requires relatively low concentration (∼0.5M) of salt for folding. These results suggest that suppression of negative charge repulsion at both active and bi-aspartate sites by salt is necessary to yield a folded protein.


Subject(s)
Protein Conformation/drug effects , Protein Folding/drug effects , Ribonuclease H/chemistry , Catalytic Domain/drug effects , Chlorides/chemistry , Chlorides/pharmacology , Halobacterium/drug effects , Magnesium Chloride/chemistry , Magnesium Chloride/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Mutation , Ribonuclease H/drug effects , Sodium Chloride/chemistry , Sodium Chloride/pharmacology
4.
AIDS ; 26(16): 1983-93, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-22555171

ABSTRACT

Ribonucleases H or RNases H are conserved and exist in almost every organism. They generate and remove RNA primers, which are required for DNA replication. RNases H hydrolyze RNA in RNA-DNA hybrids. RNases H and related enzymes contribute to reduction of gene expression in antisense and small-interfering RNA mechanisms for gene silencing. Retroviruses code for RNases H, which are required for DNA provirus synthesis. Their RNase H is fused to the reverse transcriptase and essential for virus replication inside the cell. Retroviruses code for four enzymes, three of which have been targeted by antiretroviral therapies. A drug against the fourth one, the retroviral RNase H, does not yet exist. The viral but not cellular RNases H should be targeted by drug design. Some details will be discussed here. Furthermore, a compound is described, which enables the RNase H to kill cell-free HIV particles by driving the virus into suicide - with potential use as a microbicide.


Subject(s)
Anti-Infective Agents/pharmacology , HIV Reverse Transcriptase/genetics , HIV-1/genetics , RNA, Viral/drug effects , Ribonuclease H/genetics , Virus Replication/genetics , Amino Acid Sequence , DNA Replication , Drug Design , Gene Expression Regulation, Viral/drug effects , Gene Silencing , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/enzymology , Humans , Mutation , Nucleic Acid Amplification Techniques , Ribonuclease H/drug effects , Ribonuclease H/metabolism , Virus Replication/drug effects
5.
J Med Virol ; 79(3): 207-11, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17245724

ABSTRACT

Recent studies have highlighted the need of investigating the in vivo role of ribonuclease H (RNase H) in nucleoside reverse transcriptase inhibitors (NRTIs) resistance. The prevalence of RNase H mutations in naive and in NRTI pre-treated patients in regimen failure were compared and some specific associations between NRTI resistance mutations and RNase H mutations were determined. Four positions were mutated more frequently in pre-treated patients than in naive patients: L469T/I/M/H, T470P/S/E/K, A554T/L/K, and K558R/G/E. Mutations at position K558 were also associated with the number of thymidine analog mutations (TAMs). These results suggest that these mutations could play a role in NRTI resistance.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV Infections/virology , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Mutation, Missense , Reverse Transcriptase Inhibitors/pharmacology , Amino Acid Substitution/genetics , HIV Infections/drug therapy , HIV Reverse Transcriptase/drug effects , HIV-1/genetics , Humans , Models, Molecular , Protein Subunits/drug effects , Protein Subunits/genetics , Ribonuclease H/drug effects , Ribonuclease H/genetics
6.
Mol Cancer Ther ; 5(7): 1683-92, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891454

ABSTRACT

Chronic myeloid leukemia (CML) develops when a hematopoietic stem cell acquires the Philadelphia chromosome carrying the BCR/ABL fusion gene. This gives the transformed cells a proliferative advantage over normal hematopoietic cells. Silencing the BCR/ABL oncogene by treatment with specific drugs remains an important therapeutic goal. In this work, we used locked nucleic acid (LNA)-modified oligonucleotides to silence BCR/ABL and reduce CML cell proliferation, as these oligonucleotides are resistant to nucleases and exhibit an exceptional affinity for cognate RNA. The anti-BCR/ABL oligonucleotides were designed as LNA-DNA gapmers, consisting of end blocks of 3/4 LNA monomers and a central DNA stretch of 13/14 deoxyribonucleotides. The gapmers were complementary to the b2a2 and b3a2 mRNA junctions with which they form hybrid duplexes that have melting temperatures of 79 degrees C and 75 degrees C, respectively, in a 20 mmol/L NaCl-buffered (pH 7.4) solution. Like DNA, the designed LNA-DNA gapmers were capable of activating RNase H and promote cleavage of the target b2a2 and b3a2 BCR/ABL mRNAs. The treatment of CML cells with junction-specific antisense gapmers resulted in a strong and specific reduction of the levels of BCR/ABL transcripts ( approximately 20% of control) and protein p210(BCR/ABL) ( approximately 30% of control). Moreover, the antisense oligonucleotides suppressed cell growth up to 40% of control and induced apoptosis, as indicated by the increase of caspase-3/7 activity in the treated cells. Finally, the b2a2-specific antisense gapmer used in combination with STI571 (imatinib mesylate), a tyrosine kinase inhibitor of p210(BCR/ABL), produced an enhanced antiproliferative effect in KYO-1 cells, which compared with K562 cells are refractory to STI571. The data of this study support the application of BCR/ABL antisense LNA-DNA gapmers, used either alone or in combination with STI571, as potential antileukemic agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Oligonucleotides, Antisense/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/chemistry , Apoptosis/genetics , Benzamides , Cell Proliferation/drug effects , Fusion Proteins, bcr-abl/analysis , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Oligonucleotides , Oligonucleotides, Antisense/chemistry , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , RNA, Messenger/antagonists & inhibitors , Ribonuclease H/drug effects , Transcription, Genetic/drug effects , Tumor Cells, Cultured
7.
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
8.
Phytother Res ; 16 Suppl 1: S57-62, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11933141

ABSTRACT

In an effort to develop new drugs preventing the growth of human immunodeficiency virus (HIV), we developed an in vitro assay method of ribonuclease H (RNase H) activity associated with reverse transcriptase (RT) from HIV-1. Some naphthoquinones, such as 1,4-naphthoquinone (1), vitamin K(3) (2), juglone (3) and plumbagin (6), moderately inhibited RNase H activity, and others, including naphthazarin (5) and shikonins (8-9, 18-23), showed weak inhibition. Diterpenoid quinones, tanshinones (24-28), had also moderate inhibition against RNase H activity. Of these quinones, compound 1 showed the most potent inhibition on RNase H activity with a 50% inhibitory concentration (IC(50)) of 9.5 microM, together with moderate inhibition against RNA-dependent and DNA-dependent DNA polymerase (RDDP and DDDP) activities with IC(50) values of 69 and 36 microM, respectively. Compounds 3 and 5 showed significant inhibition against RDDP (IC(50) = 8 and 10 microM, respectively) and DDDP (IC(50) = 5 and 7 microM, respectively) activities. The structure-activity relationship of the naphthoquinones suggested that non-hydroxylated naphthoquinones (1 and 2) showed significant inhibition of RNase H activity, whereas 5-hydroxylated naphthoquinones (3 and 5) showed potent inhibition against RDDP and DDDP activities.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV Reverse Transcriptase/drug effects , HIV-1/drug effects , Phytotherapy , Quinones/pharmacology , Ribonuclease H/drug effects , Dose-Response Relationship, Drug , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , HIV-1/enzymology , Humans , Inhibitory Concentration 50 , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Quinones/administration & dosage , Quinones/therapeutic use
9.
Brain Res ; 916(1-2): 136-42, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11597600

ABSTRACT

PURPOSE: To determine whether antisense oligonucleotides (AODNs) targeted against CRX, a photoreceptor-specific trans-acting factor, suppress CRX expression and interphotoreceptor retinoid binding protein (IRBP) promoter activity. METHODS: Cultures of human retinoblastoma cells were transfected with chloramphenicol acetyltransferase (CAT) reporter plasmids containing a mouse IRBP promoter and AODNs directed against CRX. RT-PCR using primers specific to CRX, OTX2, GAPDH, or RNase H was conducted on total RNA isolated from retinoblastoma cells at various times following transfection with AODNs. RESULTS: Transfection of retinoblastoma cells with IRBP promoter CAT constructs alone produced high activity. Co-transfection with AODNs suppressed IRBP promoter activity in a concentration-dependent manner, with half-maximal effect produced at about 2 nM AODN concentration. Transfection with CAT constructs containing an SV40 promoter produced high activity that was unaffected by co-transfection with AODNs. RT-PCR products were obtained for all target sequences. CRX RT-PCR product from cells transfected with AODNs was greatly diminished following transfection with an AODN whereas control transcripts, including that of OTX2, were relatively unaffected. CONCLUSIONS: The CRX-specific AODNs specifically and potently suppressed CRX expression and IRBP promoter activity, as measured by RT-PCR and transient transfection assays, respectively. Little or no effect was seen on controls. These data suggest that endogenous CRX is required for IRBP promoter activity in retinoblastoma cells.


Subject(s)
Eye Proteins , Gene Expression Regulation/physiology , Homeodomain Proteins/drug effects , Oligonucleotides, Antisense/pharmacology , Promoter Regions, Genetic/drug effects , Retina/drug effects , Retinol-Binding Proteins/drug effects , Trans-Activators/drug effects , Transcription, Genetic/drug effects , Base Sequence/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Genes, Reporter/physiology , Genetic Vectors/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Homeodomain Proteins/genetics , Humans , Molecular Sequence Data , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Otx Transcription Factors , Promoter Regions, Genetic/physiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Retina/metabolism , Retinoblastoma , Retinol-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease H/drug effects , Ribonuclease H/genetics , Trans-Activators/genetics , Transcription, Genetic/genetics , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
10.
Proc Natl Acad Sci U S A ; 97(10): 5633-8, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10805816

ABSTRACT

Insufficient efficacy and/or specificity of antisense oligonucleotides limit their in vivo usefulness. We demonstrate here that a high-affinity DNA analog, locked nucleic acid (LNA), confers several desired properties to antisense agents. Unlike DNA, LNA/DNA copolymers were not degraded readily in blood serum and cell extracts. However, like DNA, the LNA/DNA copolymers were capable of activating RNase H, an important antisense mechanism of action. In contrast to phosphorothioate-containing oligonucleotides, isosequential LNA analogs did not cause detectable toxic reactions in rat brain. LNA/DNA copolymers exhibited potent antisense activity on assay systems as disparate as a G-protein-coupled receptor in living rat brain and an Escherichia coli reporter gene. LNA-containing oligonucleotides will likely be useful for many antisense applications.


Subject(s)
Oligodeoxyribonucleotides, Antisense/chemistry , Oligodeoxyribonucleotides, Antisense/pharmacology , Animals , Base Sequence , Biological Transport , Breast Neoplasms , Caudate Nucleus/drug effects , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiology , Drug Design , Enzyme Activation , Female , Humans , Oligodeoxyribonucleotides, Antisense/pharmacokinetics , Putamen/drug effects , Rats , Receptors, Opioid, delta/genetics , Ribonuclease H/drug effects , Ribonuclease H/metabolism , Stereotaxic Techniques , Thionucleotides , Tumor Cells, Cultured
11.
Antisense Nucleic Acid Drug Dev ; 10(6): 463-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11198930

ABSTRACT

The potential pharmacologic benefits of using peptide nucleic acid (PNA) as an antisense agent are tempered by its incapacity to activate RNase H. The mixed backbone oligonucleotide (ON) (or gapmer) approach, in which a short internal window of RNAse H-competent residues is embedded within an RNase H-incompetent ON has not been applied previously to PNA because PNA and DNA hybridize to RNA with very different helical structures, creating structural perturbations at the two PNA-DNA junctions. It is demonstrated here for the first time that a short internal phosphodiester window within a PNA is sufficient to evoke the RNase H-dependent cleavage of a targeted RNA and to abrogate translation elongation in a well-characterized in vitro assay.


Subject(s)
Oligonucleotides, Antisense/pharmacology , Oligonucleotides/pharmacology , Peptide Nucleic Acids/pharmacology , RNA, Messenger/metabolism , Ribonuclease H/metabolism , Animals , Catalysis , Cell-Free System , DNA/chemistry , Dose-Response Relationship, Drug , Escherichia coli/enzymology , Gene Targeting , Oligonucleotides, Antisense/chemistry , Peptide Nucleic Acids/chemistry , Polymers/pharmacology , Protein Biosynthesis/drug effects , RNA, Messenger/drug effects , Rabbits , Ribonuclease H/drug effects , Ribonuclease H/genetics
12.
Bioorg Med Chem ; 5(6): 1037-42, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9222496

ABSTRACT

We have previously prepared ribozyme mimics and chemical nucleases from modified DNA containing pendant bipyridine and terpyridine groups. The ability of these modified DNA probes to support RNase H cleavage of complementary RNA is described. DNA/RNA duplexes were formed using DNA probes designed to deliver metal complexes via either the major groove or the minor groove of the duplex. The duplexes were treated with Escherichia coli RNase H. Modifications in the major groove produced the same RNA cleavage pattern as unmodified DNA probes. However, minor groove substituents inhibited RNA cleavage over a four-base region. Comparison was made with a DNA probe containing a 2'-OMe modification. Our results support enzyme binding in the minor groove of a DNA/RNA duplex. We do not observe cleavage directly across from the modified nucleoside. The RNA cleavage efficiency effected by RNase H and a DNA probe decreases as follows: unmodified DNA > or = C-5 modified DNA >> c2'-modified DNA > C1'-modified DNA. Results with 28-mer RNA substrates roughly parallel those obtained with a 159-mer RNA target. The differences observed between low and high MW RNA substrates can be explained by a much higher enzyme-substrate binding constant for the high MW target.


Subject(s)
DNA Probes/pharmacology , DNA/metabolism , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes/metabolism , RNA, Complementary/metabolism , Ribonuclease H/metabolism , Base Sequence , Binding Sites , DNA/chemistry , DNA Probes/chemistry , DNA Probes/metabolism , Escherichia coli/enzymology , Hydrolysis , Molecular Weight , Nucleic Acid Heteroduplexes/chemistry , RNA, Complementary/chemistry , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/drug effects
13.
Biochim Biophys Acta ; 1351(3): 249-55, 1997 Apr 10.
Article in English | MEDLINE | ID: mdl-9130587

ABSTRACT

Antisense oligonucleotides (ODNs) overlapping the stem-loop structure of the trans-activating responsive (TAR) element at the 5' end of HIV-1 and HIV-2 viral RNAs were tested for their inhibitory effect on cDNA synthesis by HIV-1 and HIV-2 reverse transcriptases (RT). Inhibition of reverse transcription is sequence-specific and enhanced by the presence of the RT-associated RNase H activity. The degree of inhibition obtained with the anti-TAR antisense is significantly higher than with other HIV-1 targeted antisense ODNs used before [1]. Gel retardation showed a stable specific complex between the 16- and 25-mer anti-TAR HIV-1 selected ODNs and the target region. No complex was observed with a non-inhibitor 22-mer anti-TAR ODN and with the corresponding control sequences. Targeting of the first stem-loop in the 5' region of HIV-2 RNA by anti-TAR ODNs inhibited very strongly reverse transcription by HIV-2 RT. The structure of the antisense and the target sequence affect annealing efficiency and hence the degree of inhibition of reverse transcription.


Subject(s)
HIV Long Terminal Repeat/drug effects , HIV-1/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , RNA-Directed DNA Polymerase/genetics , Base Sequence , DNA, Complementary/biosynthesis , DNA, Complementary/drug effects , DNA, Viral/biosynthesis , DNA, Viral/drug effects , Electrophoresis/methods , HIV Long Terminal Repeat/genetics , HIV Reverse Transcriptase , HIV-1/drug effects , Molecular Sequence Data , Nucleic Acid Heteroduplexes , Oligonucleotides, Antisense/chemistry , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA-Directed DNA Polymerase/drug effects , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H/drug effects , Ribonuclease H/genetics , Ribonuclease H/metabolism , Virus Replication/drug effects , Virus Replication/genetics
14.
Biochemistry ; 34(31): 9936-43, 1995 Aug 08.
Article in English | MEDLINE | ID: mdl-7543283

ABSTRACT

The stimulatory effect of Mg2+ and Mn2+ on the ribonuclease H (RNase H) functions of HIV-1 reverse transcriptase (RT) has been evaluated using a model 90-nt RNA template/36-nt DNA primer. Wild type enzyme exhibits similar endonuclease and directional processing activities in response to both cations, while RNase H activity (hydrolysis of double-stranded RNA) is only evident in the presence of Mn2+. Enzyme altered at the p66 residue Glu478 (Glu478-->Gln478), which participates in metal ion binding, is completely inactive in Mg2+. However, Mn2+ restores specifically its endoribonuclease activity. In the presence of Mn2+, mutant RT also catalyzes specific removal of the tRNA replication primer, eliminating the possibility of contaminating Escherichia coli RNase H in our recombinant enzyme. However, the efficiency with which mutant RT catalyzes transfer of nascent DNA between RNA templates (an event mandating RNase H activity) is severely reduced. These findings raise the possibility that directional processing activity is required to accelerate transfer of nascent DNA between templates during retroviral replication.


Subject(s)
Cations, Divalent/pharmacology , HIV-1/enzymology , RNA-Directed DNA Polymerase/metabolism , Ribonuclease H/metabolism , Base Sequence , Binding Sites , DNA, Viral/metabolism , DNA-Directed DNA Polymerase/metabolism , HIV Reverse Transcriptase , HIV-1/growth & development , Magnesium/pharmacology , Manganese/pharmacology , Molecular Sequence Data , Nucleic Acid Heteroduplexes/metabolism , RNA , RNA, Transfer/metabolism , RNA, Viral/metabolism , RNA-Directed DNA Polymerase/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Ribonuclease H/drug effects , Substrate Specificity
15.
Biochem Biophys Res Commun ; 211(3): 1041-6, 1995 Jun 26.
Article in English | MEDLINE | ID: mdl-7541196

ABSTRACT

We demonstrate the degradation of RNA bound to an antisense oligonucleotide by a reverse transcriptase enzyme-associated RNase H activity. We found that phosphorothioate oligonucleotides inhibit the RNase H activity by binding to AMV RT, rather than to the template RNA, whereas the RNase H activity of HIV-1 RT is not affected by the antisense phosphorothioate oligonucleotide. Selective inhibition of HIV-1 gene expression involves the degradation of the template RNA bound to the antisense phosphorothioate oligonucleotide by the RNase H activity associated with the HIV-1 polymerase.


Subject(s)
HIV-1/enzymology , Oligonucleotides, Antisense/pharmacology , RNA-Directed DNA Polymerase/metabolism , Ribonuclease H/drug effects , Thionucleotides/pharmacology , Transcription, Genetic/drug effects , Avian Myeloblastosis Virus/enzymology , Base Sequence , Molecular Sequence Data , Nucleic Acid Heteroduplexes/metabolism , RNA/metabolism , Ribonuclease H/antagonists & inhibitors
16.
J Biol Chem ; 269(41): 25922-7, 1994 Oct 14.
Article in English | MEDLINE | ID: mdl-7523396

ABSTRACT

Cleavage specificity of RNase HI was examined on model Okazaki fragments, to determine the likely role of this nuclease in lagging strand DNA replication. Each substrate was prepared by annealing a short RNA primer, made by transcription in vitro, to a single-stranded synthetic DNA template, and subsequently extending the primer by DNA polymerization. The calf thymus RNase HI makes a structure-specific endonucleolytic cleavage in the RNA primer, releasing it intact, and leaving a mono-ribonucleotide at the 5' terminus of the RNA-DNA junction. This specific cleavage, one nucleotide upstream of the RNA-DNA junction, is RNA primer sequence- and length-independent. Cleavage specificity is lost if the RNA primer is not extended with DNA, or if the substrate has a nick at the RNA-DNA junction. In addition, the cleavage at a single site requires Mg2+. Cleavage in the presence of Mn2+ is less specific. Neither human immunodeficiency virus reverse transcriptase nor Escherichia coli RNases H perform such a structure-specific cleavage before an RNA-DNA junction. Our work indicates that calf RNase HI is designed to recognize Okazaki fragments. It has the specificity to remove their initiator RNA segments, except for one ribonucleotide, by a single endonucleolytic cleavage in vivo.


Subject(s)
DNA Replication , DNA/metabolism , RNA/metabolism , Ribonuclease H/metabolism , Thymus Gland/enzymology , Animals , Base Sequence , Cattle , HIV/enzymology , Magnesium/pharmacology , Manganese/pharmacology , Molecular Sequence Data , RNA-Directed DNA Polymerase/metabolism , Ribonuclease H/drug effects , Substrate Specificity
17.
J Biol Chem ; 269(34): 21741-7, 1994 Aug 26.
Article in English | MEDLINE | ID: mdl-7520442

ABSTRACT

An active p15 RNase H domain, consisting of amino acids 427-560 of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) and a genetically engineered penta-histidine N-terminal affinity tag, was expressed in Escherichia coli and purified to apparent homogeneity by immobilized metal affinity chromatography. The purified p15 RNase H domain exhibited no substrate preference for [3H]poly(rG).poly(dC) compared to [3H]poly(rA).poly(dT), in contrast with the HIV-1 RT-associated RNase H, which showed a 30-fold preference for the former substrate. Unlike the HIV-1 RT-associated RNase H, when challenged with unlabeled substrate, the recombinant p15 RNase H domain was relatively nonprocessive in RNA degradative activity of the [3H]poly(rA).poly(dT) duplex. Kinetic studies using p15 RNase H showed substrate inhibition with an apparent K(i) value of 0.12 micron for the [3H]poly(rA).poly(dT) hybrid. Substrate inhibition was not observed for the HIV-1 RT-associated RNase H. The results show that the isolated p15 HIV-1 RNase H domain is functionally distinct from the recombinant HIV-1 RT-associated RNase H.


Subject(s)
HIV-1/enzymology , Peptide Fragments/metabolism , RNA-Directed DNA Polymerase/metabolism , Ribonuclease H/metabolism , Amino Acid Sequence , Base Sequence , Cations, Divalent/pharmacology , Escherichia coli/genetics , HIV Reverse Transcriptase , Molecular Sequence Data , Peptide Fragments/genetics , Protein Engineering , RNA/metabolism , RNA-Directed DNA Polymerase/drug effects , RNA-Directed DNA Polymerase/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Inhibitors , Ribonuclease H/drug effects , Ribonuclease H/genetics , Substrate Specificity
18.
Anticancer Drug Des ; 8(1): 33-51, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8386514

ABSTRACT

A previous report has demonstrated that normal phosphodiester oligodeoxynucleotides could direct extensive non-targeted ribonuclease (RNase) H-dependent effects, and that greatly enhanced specificity could be achieved upon methylphosphonodiester substitution of terminal phosphodiester residues. In this report, we extend our previous observations to show that phosphorothioate oligodeoxynucleotides also direct substantial inappropriate RNase H-mediated hydrolysis of non-targeted RNA. Chimeric methylphosphonodiester/phosphodiesters were found to be capable of efficiently directing RNase H when the central phosphodiester section was reduced to just two contiguous internucleoside linkages. Furthermore, cleavage of non-target RNA sites was found to be undetectable, or minimal in extent, when RNase H was directed by such chimeras. In addition, we show that analogue structures which contain three, or fewer, phosphodiester residues in otherwise methylphosphonodiester molecules were imported into cells via the comparatively more efficient route taken by methylphosphonates, rather than by receptor-mediated endocytosis, which is generally characteristic for polyanionic structures. Evidence is presented that the primary process responsible for enhanced uptake is an active mechanism. Nevertheless, a proportion of the applied oligodeoxynucleotide analogues, which demonstrate augmented uptake, appear to have penetrated into the cytoplasmic cellular compartment. The present results suggest that chimeric molecules of the type we describe here may show considerable utility as antisense effectors due to their increased cellular import, access to the intracellular compartments, and their highly efficient and specific direction of RNase H.


Subject(s)
Oligonucleotides, Antisense/pharmacokinetics , Ribonuclease H/drug effects , Thionucleotides/pharmacokinetics , Transcription, Genetic/drug effects , Base Sequence , Blotting, Northern , Chromatography, High Pressure Liquid , Densitometry , Flow Cytometry , Humans , Molecular Sequence Data , Oligonucleotides, Antisense/chemical synthesis , Oligonucleotides, Antisense/pharmacology , Polymerase Chain Reaction , RNA, Neoplasm/isolation & purification , Sensitivity and Specificity , Thionucleotides/chemical synthesis , Thionucleotides/pharmacology , Tumor Cells, Cultured
19.
Biochimie ; 75(1-2): 127-34, 1993.
Article in English | MEDLINE | ID: mdl-7684932

ABSTRACT

A recombinant homodimer p66/p66 of the HIV-1 reverse transcriptase (RT) was expressed in and purified from a protease-deficient strain of the yeast Saccharomyces cerevisiae. The RNase H activity associated with the homodimer was biochemically characterized. The effect of cations and the hybrid substrate specificity were studied. Some compounds which have been found to inhibit retroviral replication were tested as potential inhibitors of the retroviral DNA polymerase and RNase H activities. Most of these compounds inhibited preferentially the DNA polymerase activity. On the other hand, only suramin was found to inhibit RNase H more efficiently than DNA polymerase. As in the case of the DNA polymerase activity, the thiol-reacting agent N-ethylmaleimide (NEM) did not affect the RNAse H activity of HIV RT. When the effect of NEM was tested against E coli RNase H, a weak inhibitory effect was detected. Surprisingly, NEM strongly inhibits the same bacterial RNase H in the presence of a recombinant form of HIV RT devoid of nuclease activity. These results strongly suggest an interaction between E coli RNase H and HIV-1 RT.


Subject(s)
Ethylmaleimide/pharmacology , HIV-1 , Nucleic Acid Synthesis Inhibitors , RNA-Directed DNA Polymerase/metabolism , Ribonuclease H/metabolism , Benzodiazepines/pharmacology , Biopolymers , HIV Reverse Transcriptase , Imidazoles/pharmacology , Phosphonoacetic Acid/pharmacology , Pyridoxal Phosphate/pharmacology , RNA-Directed DNA Polymerase/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Reverse Transcriptase Inhibitors , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/chemistry , Ribonuclease H/drug effects , Suramin/pharmacology
20.
Anticancer Drug Des ; 7(1): 37-48, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1311929

ABSTRACT

Chimeric oligodeoxynucleotides, comprised of internal phosphodiester and terminal methylphosphonodiester sections, possess many beneficial characteristics as antisense effectors. We have investigated the effects of progressive replacement of phosphodiester by methylphosphonodiester linkages on hybrid stability with complementary RNA and DNA. The melting temperatures (Tms) of oligodeoxynucleotide/RNA heteroduplexes were found to decrease dramatically with increasing methylphosphonate substitution. In contrast, a smaller reduction in Tm was observed for comparable DNA heteroduplexes. This disparate reduction in hybrid stability was found with both the G + C-rich human c-myc and A + T-rich human c-Ha-ras sequences used, suggesting that methylphosphonate oligodeoxynucleotide analogues generally hybridize with less affinity to RNA than DNA. RNase H assays were employed to determine if the noted decreases in Tm impaired the ability of chimeric oligodeoxynucleotides to direct the degradation of RNA. Contrary to expectation, increasing methylphosphonate substitution gave rise to increasing rates of RNA degradation for both the c-myc and c-Ha-ras series. The present results suggest that chimeric oligodeoxynucleotide analogues may be of considerable utility as antisense agents in systems where RNase H is thought to make a major contribution to inhibition of gene expression.


Subject(s)
Oligonucleotides, Antisense/pharmacology , Ribonuclease H/drug effects , Amino Acid Sequence , Base Sequence , Molecular Sequence Data , Oligonucleotides, Antisense/chemistry , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL