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










Publication year range
1.
Viruses ; 13(1)2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33477685

ABSTRACT

HIV reverse transcriptases (RTs) convert viral genomic RNA into double-stranded DNA. During reverse transcription, polypurine tracts (PPTs) resilient to RNase H cleavage are used as primers for plus-strand DNA synthesis. Nonnucleoside RT inhibitors (NNRTIs) can interfere with the initiation of plus-strand DNA synthesis by enhancing PPT removal, while HIV RT connection subdomain mutations N348I and N348I/T369I mitigate this effect by altering RNase H cleavage specificity. Now, we demonstrate that among approved nonnucleoside RT inhibitors (NNRTIs), nevirapine and doravirine show the largest effects. The combination N348I/T369I in HIV-1BH10 RT has a dominant effect on the RNase H cleavage specificity at the PPT/U3 site. Biochemical studies showed that wild-type HIV-1 and HIV-2 RTs were able to process efficiently and accurately all tested HIV PPT sequences. However, the cleavage accuracy at the PPT/U3 junction shown by the HIV-2EHO RT was further improved after substituting the sequence YQEPFKNLKT of HIV-1BH10 RT (positions 342-351) for the equivalent residues of the HIV-2 enzyme (HQGDKILKV). Our results highlight the role of ß-sheets 17 and 18 and their connecting loop (residues 342-350) in the connection subdomain of the large subunit, in determining the RNase H cleavage window of HIV RTs.


Subject(s)
Genome, Viral , HIV Infections/virology , HIV Long Terminal Repeat , HIV-1/physiology , RNA, Viral , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Base Sequence , HIV Infections/drug therapy , HIV-1/drug effects , Humans , Models, Molecular , Molecular Conformation , Mutagenesis , Protein Binding , Proteolysis , RNA, Viral/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/therapeutic use , Ribonuclease H, Human Immunodeficiency Virus/chemistry
2.
ACS Infect Dis ; 5(11): 1963-1974, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31577424

ABSTRACT

The ribonuclease H (RNH) activity of HIV-1 reverse transcriptase (RT) is essential for viral replication and can be a target for drug development. Yet, no RNH inhibitor to date has substantial antiviral activity to allow advancement into clinical development. Herein, we describe our characterization of the detailed binding mechanisms of RNH active-site inhibitors, YLC2-155 and ZW566, that bind to the RNH domain through divalent metal ions, using NMR, molecular docking, and quantum mechanical calculations. In the presence of Mg2+, NMR spectra of RNH exhibited split (two) resonances for some residues upon inhibitor binding, suggesting two binding modes, an observation consistent with the docking results. The relative populations of the two binding conformers were independent of inhibitor or Mg2+ concentration, with one conformation consistently more favored. In our docking study, one distinctive pose of ZW566 showed more interactions with surrounding residues of RNH compared to the analogous binding pose of YLC2-155. Inhibitor titration experiments revealed a lower dissociation constant for ZW566 compared to YLC2-155, in agreement with its higher inhibitory activity. Mg2+ titration data also indicated a stronger dependence on Mg2+ for the RNH interaction with ZW566 compared to YLC2-155. Combined docking and quantum mechanical calculation results suggest that stronger metal coordination as well as more protein-inhibitor interactions may account for the higher binding affinity of ZW566. These findings support the idea that strategies for the development of potent competitive active site RNH inhibitors should take into account not only metal-inhibitor coordination but also protein-inhibitor interaction and conformational selectivity.


Subject(s)
Anti-HIV Agents/chemistry , Enzyme Inhibitors/chemistry , HIV Infections/virology , HIV-1/enzymology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Anti-HIV Agents/metabolism , Catalytic Domain , Enzyme Inhibitors/metabolism , HIV-1/drug effects , HIV-1/genetics , Humans , Isoquinolines/chemistry , Isoquinolines/metabolism , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Molecular Docking Simulation , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism
3.
Eur J Med Chem ; 166: 390-399, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30739822

ABSTRACT

The pharmacophore of active site inhibitors of human immunodeficiency virus (HIV) reverse transcriptase (RT)-associated RNase H typically entails a flexible linker connecting the chelating core and the hydrophobic aromatics. We report herein that novel 3-hydroxypyrimidine-2,4-dione (HPD) subtypes with a nonflexible C-6 carbonyl linkage exhibited potent and selective biochemical inhibitory profiles with strong RNase H inhibition at low nM, weak to moderate integrase strand transfer (INST) inhibition at low µM, and no to marginal RT polymerase (pol) inhibition up to 10 µM. A few analogues also demonstrated significant antiviral activity without cytotoxicity. The overall inhibitory profile is comparable to or better than that of previous HPD subtypes with a flexible C-6 linker, suggesting that the nonflexible carbonyl linker can be tolerated in the design of novel HIV RNase H active site inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Catalytic Domain , Drug Design , Enzyme Inhibitors/metabolism , HIV-1/drug effects , Inhibitory Concentration 50 , Molecular Docking Simulation , Pyrimidinones/metabolism , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism
4.
Eur J Med Chem ; 156: 680-691, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30031978

ABSTRACT

Human immunodeficiency virus (HIV) reverse transcriptase (RT)-associated ribonuclease H (RNase H) remains an unvalidated drug target. Reported HIV RNase H inhibitors generally lack significant antiviral activity. We report herein the design, synthesis, biochemical and antiviral evaluations of a new 6-biphenylmethyl subtype of the 3-hydroxypyrimidine-2,4-dione (HPD) chemotype. In biochemical assays, analogues of this new subtype potently inhibited RT RNase H in low nanomolar range without inhibiting RT polymerase (pol) or integrase strand transfer (INST) at the highest concentrations tested. In cell-based assays, a few analogues inhibited HIV in low micromolar range without cytotoxicity at concentrations up to 100 µM.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV-1/drug effects , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Catalytic Domain/drug effects , Cell Line , Drug Design , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/enzymology , Humans , Methylation , Models, Molecular , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship
5.
Viruses ; 9(11)2017 11 08.
Article in English | MEDLINE | ID: mdl-29117130

ABSTRACT

The South African national treatment programme includes nucleoside reverse transcriptase inhibitors (NRTIs) in both first and second line highly active antiretroviral therapy regimens. Mutations in the RNase H domain have been associated with resistance to NRTIs but primarily in HIV-1 subtype B studies. Here, we investigated the prevalence and association of RNase H mutations with NRTI resistance in sequences from HIV-1 subtype C infected individuals. RNase H sequences from 112 NRTI treated but virologically failing individuals and 28 antiretroviral therapy (ART)-naive individuals were generated and analysed. In addition, sequences from 359 subtype C ART-naive sequences were downloaded from Los Alamos database to give a total of 387 sequences from ART-naive individuals for the analysis. Fisher's exact test was used to identify mutations and Bayesian network learning was applied to identify novel NRTI resistance mutation pathways in RNase H domain. The mutations A435L, S468A, T470S, L484I, A508S, Q509L, L517I, Q524E and E529D were more prevalent in sequences from treatment-experienced compared to antiretroviral treatment naive individuals, however, only the E529D mutation remained significant after correction for multiple comparison. Our findings suggest a potential interaction between E529D and NRTI-treatment; however, site-directed mutagenesis is needed to understand the impact of this RNase H mutation.


Subject(s)
HIV-1/genetics , Mutation , Reverse Transcriptase Inhibitors/therapeutic use , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/genetics , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Bayes Theorem , Databases, Genetic , Drug Resistance, Viral/genetics , Genotype , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Humans , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Sequence Analysis, DNA
6.
Biochemistry ; 56(27): 3434-3442, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28627879

ABSTRACT

The heterodimeric human immunodeficiency virus type 1 reverse transcriptase is composed of p66 and p51 subunits. While in the p51 subunit, the connection domain is tucked in the polymerase cleft; it is effectively displaced from the cleft of the catalytically active p66 subunit. How is the connection domain relocated from the polymerase cleft of p66? Does the RNase H domain have any role in this process? To answer this question, we extended the C-terminal region of p51 by stepwise addition of N-terminal motifs of RNase H domain to generate p54, p57, p60, and p63 derivatives. We found all of the C-terminal extended derivatives of p51 assume open conformation, bind to the template-primer, and catalyze the polymerase reaction. Glycerol gradient ultracentrifugation analysis showed that only p54 sedimented as a monomer, while other derivatives were in a homodimeric conformation. We proposed a model to explain the monomeric conformation of catalytically active p54 derivative carrying additional 21-residues long ß1'-ß2' motif from the RNase H domain. Our results indicate that the ß1'-ß2' motif of the RNase H domain may be responsible for displacing the connection domain from the polymerase cleft of putative monomeric p66. The unstable elongated p66 molecule may then readily dimerize with p51 to assume a stable dimeric conformation.


Subject(s)
HIV Reverse Transcriptase/chemistry , HIV-1/enzymology , Models, Molecular , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Amino Acid Motifs , Dimerization , Enzyme Stability , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Docking Simulation , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism
7.
Curr Med Chem ; 21(17): 1956-67, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24438523

ABSTRACT

Reverse transcription of human immunodeficiency virus type 1 (HIV-1) is a crucial step in the life cycle initiated by the viral-coded reverse transcriptase (RT), functioning as RNA- and DNA-dependent DNA polymerase (RDDP and DDDP) and the ribonuclease H (RNase H). The RNase H functions to degrade the RNA strand of the RNA:DNA heteroduplex, which makes it an attractive target for rational anti-HIV-1 drug design and development. Although development of drugs targeting the DNA polymerase have been highly successful, the discovery of drugable inhibitors of HIV RNase H is still in its infancy and none of RNase H inhibitors has reached the clinical development stage currently. This review describes the recent progress in the HIV-1 RNase H inhibitors, focusing on their chemical feature, mechanism and the structure-activity relationship (SAR).


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , HIV-1/enzymology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Drug Design , HIV Infections/enzymology , HIV Infections/virology , HIV-1/drug effects , Humans , Models, Molecular , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship
8.
Methods Mol Biol ; 1087: 71-84, 2014.
Article in English | MEDLINE | ID: mdl-24158815

ABSTRACT

The Ribonuclease (RNase) H is one of the four enzymes encoded by all retroviruses, including HIV. Its main activity is the hydrolysis of the RNA moiety in RNA-DNA hybrids. The RNase H ribonuclease is essential in the retroviral life cycle, since it generates and removes primers needed by the Reverse Transcriptase (RT) for initiation of DNA synthesis. Retroviruses lacking RNase H activity are noninfectious. Despite its importance, RNase H is the only enzyme of HIV not yet targeted by antiretroviral therapy. Here, we describe functions and mechanisms of RNase H during the HIV life cycle and describe a cleavage assay, which is suitable to determine RNase H activity in samples of various kinds. In this assay, an artificial, fluorescence-labeled RNA-DNA hybrid is cleaved in vitro by an RT/RNase H enzyme. Cleavage products are analyzed by denaturing polyacrylamide gel electrophoresis (PAGE). This assay may be used to detect the RNase H, assess the effect of inhibitors, or even activators, of the RNase H, as we have described, as candidates for novel antiretroviral agents.


Subject(s)
Anti-HIV Agents/pharmacology , Enzyme Assays/methods , HIV-1/enzymology , Ribonuclease H, Human Immunodeficiency Virus/metabolism , HIV-1/drug effects , HIV-1/physiology , Models, Molecular , Protein Denaturation , Protein Structure, Secondary , Proteolysis , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Substrate Specificity
9.
Biochemistry ; 52(51): 9318-28, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24303887

ABSTRACT

At high temperatures, RNA denaturation can improve the efficiency and specificity of reverse transcription. Refined structures and molecular models of HIV-1 reverse transcriptases (RTs) from phylogenetically distant clades (i.e., group M subtype B and group O) revealed a major interaction between the template-primer and the Arg³58-Gly³59-Ala³6° triad in the large subunit of HIV-1M/B RT. However, fewer contacts were predicted for the equivalent Lys³58-Ala³59-Ser³6° triad of HIV-1O RT and the nucleic acid. An engineered HIV-1O K358R/A359G/S360A RT showed increased cDNA synthesis efficiency above 68 °C, as determined by qualitative and quantitative reverse transcription polymerase chain reactions. In comparison with wild-type HIV-1O RT, the mutant enzyme showed higher thermal stability but retained wild-type RNase H activity. Mutations that increased the accuracy of HIV-1M/B RTs were tested in combination with the K358R/A359G/S360A triple mutation. Some of them (e.g., F61A, K65R, K65R/V75I, and V148I) had a negative effect on reverse transcription efficiency above 65 °C. RTs with improved DNA binding affinities also showed higher cDNA synthesis efficiencies at elevated temperatures. Two of the most thermostable RTs (i.e., mutants T69SSG/K358R/A359G/S360A and K358R/A359G/S360A/E478Q) showed moderately increased fidelity in forward mutation assays. Our results demonstrate that the triad of Arg³58, Gly³59, and Ala³6° in the major groove binding track of HIV-1 RT is a major target for RT stabilization, and most relevant for improving reverse transcription efficiency at high temperatures.


Subject(s)
DNA, Complementary/biosynthesis , DNA/metabolism , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Models, Molecular , RNA, Viral/metabolism , Amino Acid Motifs , Amino Acid Substitution , Binding Sites , Computer Simulation , DNA/chemistry , Databases, Protein , Enzyme Stability , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV-1/metabolism , Hot Temperature , Kinetics , Molecular Conformation , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , RNA, Viral/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Thymine Nucleotides/chemistry , Thymine Nucleotides/metabolism
10.
J Med Chem ; 56(13): 5436-45, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23631411

ABSTRACT

The thienopyrimidinone 5,6-dimethyl-2-(4-nitrophenyl)thieno[2,3-d]pyrimidin-4(3H)-one (DNTP) occupies the interface between the p66 ribonuclease H (RNase H) domain and p51 thumb of human immunodeficiency virus reverse transcriptase (HIV RT), thereby inducing a conformational change incompatible with catalysis. Here, we combined biochemical characterization of 39 DNTP derivatives with antiviral testing of selected compounds. In addition to wild-type HIV-1 RT, derivatives were evaluated with rationally designed, p66/p51 heterodimers exhibiting high-level DNTP sensitivity or resistance. This strategy identified 3',4'-dihydroxyphenyl (catechol) substituted thienopyrimidinones with submicromolar in vitro activity against both wild type HIV-1 RT and drug-resistant variants. Thermal shift analysis indicates that, in contrast to active site RNase H inhibitors, these thienopyrimidinones destabilize the enzyme, in some instances reducing the Tm by 5 °C. Importantly, catechol-containing thienopyrimidinones also inhibit HIV-1 replication in cells. Our data strengthen the case for allosteric inhibition of HIV RNase H activity, providing a platform for designing improved antagonists for use in combination antiviral therapy.


Subject(s)
Drug Resistance, Viral/drug effects , Enzyme Inhibitors/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , Pyrimidinones/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Enzyme Inhibitors/chemistry , Enzyme Stability/drug effects , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , HIV-1/enzymology , HIV-1/genetics , Humans , Models, Molecular , Molecular Structure , Mutation , Protein Binding , Protein Structure, Tertiary , Pyrimidinones/chemistry , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/genetics , Temperature
11.
Nucleic Acids Res ; 41(8): 4601-12, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23444139

ABSTRACT

Asp(443) and Glu(478) are essential active site residues in the RNase H domain of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT). We have investigated the effects of substituting Asn for Asp(443) or Gln for Glu(478) on the fidelity of DNA-dependent DNA synthesis of phylogenetically diverse HIV-1 RTs. In M13mp2 lacZα-based forward mutation assays, HIV-1 group M (BH10) and group O RTs bearing substitutions D443N, E478Q, V75I/D443N or V75I/E478Q showed 2.0- to 6.6-fold increased accuracy in comparison with the corresponding wild-type enzymes. This was a consequence of their lower base substitution error rates. One-nucleotide deletions and insertions represented between 30 and 68% of all errors identified in the mutational spectra of RNase H-deficient HIV-1 group O RTs. In comparison with the wild-type RT, these enzymes showed higher frameshift error rates and higher dissociation rate constants (koff) for DNA/DNA template-primers. The effects on frameshift fidelity were similar to those reported for mutation E89G and suggest that in HIV-1 group O RT, RNase H inactivation could affect template/primer slippage. Our results support a role for the RNase H domain during plus-strand DNA polymerization and suggest that mutations affecting RNase H function could also contribute to retrovirus variability during the later steps of reverse transcription.


Subject(s)
DNA/biosynthesis , HIV Reverse Transcriptase/metabolism , Ribonuclease H, Human Immunodeficiency Virus/genetics , Amino Acid Substitution , DNA Primers/metabolism , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , Mutation , Protein Structure, Tertiary , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Templates, Genetic
12.
Nucleic Acids Res ; 40(20): 10543-53, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22941642

ABSTRACT

HIV-1 reverse transcriptase (RT) contains a C-terminal ribonuclease H (RH) domain on its p66 subunit that can be expressed as a stable, although inactive protein. Recent studies of several RH enzymes demonstrate that substrate binding plays a major role in the creation of the active site. In the absence of substrate, the C-terminal helix E of the RT RNase H domain is dynamic, characterized by severe exchange broadening of its backbone amide resonances, so that the solution characterization of this region of the protein has been limited. Nuclear magnetic resonance studies of 13C-labeled RH as a function of experimental conditions reveal that the δ1 methyl resonance of Ile556, located in a short, random coil segment following helix E, experiences a large 13C shift corresponding to a conformational change of Ile556 that results from packing of helix E against the central ß-sheet. This shift provides a useful basis for monitoring the effects of various ligands on active site formation. Additionally, we report that the RNase H complexes formed with one or both divalent ions can be individually observed and characterized using diamagnetic Zn2+ as a substitute for Mg2+. Ordering of helix E results specifically from the interaction with the lower affinity binding to the A divalent ion site.


Subject(s)
Isoleucine/chemistry , Metals/chemistry , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Arginine/chemistry , Biocatalysis , Catalytic Domain , Cations, Divalent , Enzyme Inhibitors/chemistry , HIV Reverse Transcriptase/chemistry , Isoquinolines/chemistry , Ligands , Magnesium/chemistry , Models, Molecular , Protein Structure, Tertiary , Zinc/chemistry
13.
J Biol Chem ; 287(6): 4066-75, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22105069

ABSTRACT

The vinylogous urea, NSC727447, was proposed to allosterically inhibit ribonuclease H (RNase H) activity of human immunodeficiency virus type 1 reverse transcriptase (HIV-1 RT) by interacting with the thumb subdomain of its non-catalytic p51 subunit. Proximity of the p51 thumb to the p66 RNase H domain implied that inhibitor binding altered active site geometry, whereas protein footprinting suggested a contribution from α-helix I residues Cys-280 and Lys-281. To more thoroughly characterize the vinylogous urea binding site, horizontal alanine scanning mutagenesis between p51 residues Lys-275 and Thr-286 (comprising α-helix I and portions of the neighboring αH/αI and αI/αJ connecting loops) was combined with a limited vertical scan of Cys-280. A contribution from Cys-280 was strengthened by our observation that all substitutions at this position rendered selectively mutated, reconstituted p66/p51 heterodimers ∼45-fold less sensitive to inhibition. An ∼19-fold reduced IC(50) for p51 mutant T286A coupled with a 2-8-fold increased IC(50) when intervening residues were substituted supports our original proposal of p51 α-helix I as the vinylogous urea binding site. In contrast to these allosteric inhibitors, mutant enzymes retained equivalent sensitivity to the natural product α-hydroxytropolone inhibitor manicol, which x-ray crystallography has demonstrated functions by chelating divalent metal at the p66 RNase H active site. Finally, reduced DNA strand-transfer activity together with increased vinylogous urea sensitivity of p66/p51 heterodimers containing short p51 C-terminal deletions suggests an additional role for the p51 C terminus in nucleic acid binding that is compromised by inhibitor binding.


Subject(s)
Enzyme Inhibitors/chemistry , HIV-1/enzymology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Thiophenes/chemistry , Amino Acid Substitution , Cell Line , Crystallography, X-Ray , HIV-1/genetics , Humans , Mutagenesis , Mutation, Missense , Protein Structure, Secondary , Protein Structure, Tertiary , Ribonuclease H, Human Immunodeficiency Virus/genetics
14.
J Biol Chem ; 286(47): 40433-42, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21953456

ABSTRACT

Several physiologically relevant cations including Ca(2+), Mn(2+), and Zn(2+) have been shown to inhibit HIV reverse transcriptase (RT), presumably by competitively displacing one or more Mg(2+) ions bound to RT. We analyzed the effects of Zn(2+) on reverse transcription and compared them to Ca(2+) and Mn(2+). Using nucleotide extension efficiency as a readout, Zn(2+) showed significant inhibition of reactions with 2 mM Mg(2+), even when present at only ∼5 µM. Mn(2+) and Ca(2+) were also inhibitory but at higher concentrations. Both Mn(2+) and Zn(2+) (but not Ca(2+)) supported RT incorporation in the absence of Mg(2+) with Mn(2+) being much more efficient. The maximum extension rates with Zn(2+), Mn(2+), and Mg(2+) were ∼0.1, 1, and 3.5 nucleotides per second, respectively. Zinc supported optimal RNase H activity at ∼25 µM, similar to the optimal for nucleotide addition in the presence of low dNTP concentrations. Surprisingly, processivity (average number of nucleotides incorporated in a single binding event with enzyme) during reverse transcription was comparable with Zn(2+) and Mg(2+), and single RT molecules were able to continue extension in the presence of Zn(2+) for several hours on the same template. Consistent with this result, the half-life for RT-Zn(2+)-(primer-template) complexes was 220 ± 60 min and only 1.7 ± 1 min with Mg(2+), indicating ∼130-fold more stable binding with Zn(2+). Essentially, the presence of Zn(2+) promotes the formation of a highly stable slowly progressing RT-(primer-template) complex.


Subject(s)
Biocatalysis/drug effects , DNA Primers/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Reverse Transcriptase Inhibitors/pharmacology , Zinc/pharmacology , Avian Myeloblastosis Virus/enzymology , Calcium/pharmacology , Deoxyribonucleotides/metabolism , Dose-Response Relationship, Drug , Enzyme Stability/drug effects , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , HIV-1/genetics , HIV-1/physiology , Kinetics , Magnesium/pharmacology , Moloney murine leukemia virus/enzymology , Mutation , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Templates, Genetic , Virus Replication/drug effects
15.
J Med Chem ; 54(13): 4462-73, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21568335

ABSTRACT

The α-hydroxytroplone, manicol (5,7-dihydroxy-2-isopropenyl-9-methyl-1,2,3,4-tetrahydro-benzocyclohepten-6-one), potently and specifically inhibits ribonuclease H (RNase H) activity of human immunodeficiency virus reverse transcriptase (HIV RT) in vitro. However, manicol was ineffective in reducing virus replication in culture. Ongoing efforts to improve the potency and specificity over the lead compound led us to synthesize 14 manicol derivatives that retain the divalent metal-chelating α-hydroxytropolone pharmacophore. These efforts were augmented by a high resolution structure of p66/p51 HIV-1 RT containing the nonnucleoside reverse transcriptase inhibitor (NNRTI), TMC278 and manicol in the DNA polymerase and RNase H active sites, respectively. We demonstrate here that several modified α-hydroxytropolones exhibit antiviral activity at noncytotoxic concentrations. Inclusion of RNase H active site mutants indicated that manicol analogues can occupy an additional site in or around the DNA polymerase catalytic center. Collectively, our studies will promote future structure-based design of improved α-hydroxytropolones to complement the NRTI and NNRTI currently in clinical use.


Subject(s)
Anti-HIV Agents/chemical synthesis , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Tropolone/analogs & derivatives , Tropolone/chemical synthesis , Anti-HIV Agents/pharmacology , Benzocycloheptenes/chemistry , Catalytic Domain , Cations, Divalent , Cell Line , Coordination Complexes/chemistry , Crystallography, X-Ray , DNA-Directed DNA Polymerase/chemistry , HIV Reverse Transcriptase/chemistry , HIV-1/physiology , Humans , Manganese/chemistry , Models, Molecular , Molecular Structure , Mutation , Nitriles/chemistry , Protein Conformation , Pyrimidines/chemistry , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/genetics , Rilpivirine , Structure-Activity Relationship , Tropolone/pharmacology , Virus Replication
16.
Antimicrob Agents Chemother ; 54(9): 3913-21, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20547794

ABSTRACT

Vinylogous ureas 2-amino-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxamide and N-[3-(aminocarbonyl)-4,5-dimethyl-2-thienyl]-2-furancarboxamide (compounds 1 and 2, respectively) were recently identified to be modestly potent inhibitors of the RNase H activity of HIV-1 and HIV-2 reverse transcriptase (RT). Both compounds shared a 3-CONH(2)-substituted thiophene ring but were otherwise structurally unrelated, which prevented a precise definition of the pharmacophore. We have therefore examined a larger series of vinylogous ureas carrying amide, amine, and cycloalkane modifications of the thiophene ring of compound 1. While cycloheptane- and cyclohexane-substituted derivatives retained potency, cyclopentane and cyclooctane substitutions eliminated activity. In the presence of a cycloheptane ring, modifying the 2-NH(2) or 3-CONH(2) functions decreased the potency. With respect to compound 2, vinylogous ureas whose dimethylthiophene ring contained modifications of the 2-NH(2) and 3-CONH(2) functions were investigated. 2-NH(2)-modified analogs displayed potency equivalent to or enhanced over that of compound 2, the most active of which, compound 16, reflected intramolecular cyclization of the 2-NH(2) and 3-CONH(2) groups. Molecular modeling was used to define an inhibitor binding site in the p51 thumb subdomain, suggesting that an interaction with the catalytically conserved His539 of the p66 RNase H domain could underlie inhibition of RNase H activity. Collectively, our data indicate that multiple functional groups of vinylogous ureas contribute to their potencies as RNase H inhibitors. Finally, single-molecule spectroscopy indicates that vinylogous ureas have the property of altering the reverse transcriptase orientation on a model RNA-DNA hybrid mimicking initiation plus-strand DNA synthesis.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Catalytic Domain , Humans , Models, Molecular , Molecular Structure , Temperature , Thermodynamics
17.
J Virol ; 84(15): 7625-33, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20484498

ABSTRACT

HIV/AIDS continues to be a menace to public health. Several drugs currently on the market have successfully improved the ability to manage the viral burden in infected patients. However, new drugs are needed to combat the rapid emergence of mutated forms of the virus that are resistant to existing therapies. Currently, approved drugs target three of the four major enzyme activities encoded by the virus that are critical to the HIV life cycle. Although a number of inhibitors of HIV RNase H activity have been reported, few inhibit by directly engaging the RNase H active site. Here, we describe structures of naphthyridinone-containing inhibitors bound to the RNase H active site. This class of compounds binds to the active site via two metal ions that are coordinated by catalytic site residues, D443, E478, D498, and D549. The directionality of the naphthyridinone pharmacophore is restricted by the ordering of D549 and H539 in the RNase H domain. In addition, one of the naphthyridinone-based compounds was found to bind at a second site close to the polymerase active site and non-nucleoside/nucleotide inhibitor sites in a metal-independent manner. Further characterization, using fluorescence-based thermal denaturation and a crystal structure of the isolated RNase H domain reveals that this compound can also bind the RNase H site and retains the metal-dependent binding mode of this class of molecules. These structures provide a means for structurally guided design of novel RNase H inhibitors.


Subject(s)
Enzyme Inhibitors/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/chemistry , HIV-1/drug effects , Naphthyridines/metabolism , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Binding Sites , Catalytic Domain , Cations/metabolism , Crystallography, X-Ray , HIV , HIV Reverse Transcriptase/metabolism , HIV-1/chemistry , Humans , Metals/metabolism , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Ribonuclease H, Human Immunodeficiency Virus/metabolism
18.
J Med Chem ; 52(5): 1380-7, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19178289

ABSTRACT

The RNase H activity associated with human immunodeficiency virus type 1 (HIV-1) is an attractive target for an antiretroviral drug development. We screened 20000 small-molecular-weight compounds for RNase H inhibitors and identified a novel RNase H-inhibiting structure characterized by a 5-nitro-furan-2-carboxylic acid carbamoylmethyl ester (NACME) moiety. Two NACME derivatives, 5-nitro-furan-2-carboxylic acid adamantan-1-carbamoylmethyl ester (compound 1) and 5-nitro-furan-2-carboxylic acid [[4-(4-bromo-phenyl)-thiazol-2-yl]-(tetrahydro-furan-2-ylmethyl)-carbamoyl]-methyl ester (compound 2), effectively blocked HIV-1 and MLV RT-associated RNase H activities with IC(50)s of 3-30 microM but had little effect on bacterial RNase H activity in vitro. Additionally, 20-25 microM compound 2 effectively inhibited HIV-1 replication. An in silico docking simulation indicated that the conserved His539 residue, and two metal ions in the RNase H catalytic center are involved in RNase H inhibition by NACME derivatives. Taken together, these data suggest that NACME derivatives may be potent lead compounds for development of a novel class of antiretroviral drugs.


Subject(s)
Anti-HIV Agents/chemical synthesis , Furans/chemical synthesis , HIV-1/drug effects , Reverse Transcriptase Inhibitors/chemical synthesis , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Animals , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Cell Line , Crystallography, X-Ray , Databases, Factual , Furans/chemistry , Furans/pharmacology , HIV-1/enzymology , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Structure-Activity Relationship , Virus Replication/drug effects
19.
ACS Chem Biol ; 3(10): 635-44, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18831589

ABSTRACT

High-throughput screening of National Cancer Institute libraries of synthetic and natural compounds identified the vinylogous ureas 2-amino-5,6,7,8-tetrahydro-4 H-cyclohepta[ b]thiophene-3-carboxamide (NSC727447) and N-[3-(aminocarbonyl)-4,5-dimethyl-2-thienyl]-2-furancarboxamide (NSC727448) as inhibitors of the ribonuclease H (RNase H) activity of HIV-1 and HIV-2 reverse transcriptase (RT). A Yonetani-Theorell analysis demonstrated that NSC727447, and the active-site hydroxytropolone RNase H inhibitor beta-thujaplicinol were mutually exclusive in their interaction with the RNase H domain. Mass spectrometric protein footprinting of the NSC727447 binding site indicated that residues Cys280 and Lys281 in helix I of the thumb subdomain of p51 were affected by ligand binding. Although DNA polymerase and pyrophosphorolysis activities of HIV-1 RT were less sensitive to inhibition by NSC727447, protein footprinting indicated that NSC727447 occupied the equivalent region of the p66 thumb. Site-directed mutagenesis using reconstituted p66/p51 heterodimers substituted with natural or non-natural amino acids indicates that altering the p66 RNase H primer grip significantly affects inhibitor sensitivity. NSC727447 thus represents a novel class of RNase H antagonists with a mechanism of action differing from active site, divalent metal-chelating inhibitors that have been reported.


Subject(s)
Furans/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Thiophenes/pharmacology , Urea/analogs & derivatives , Binding Sites , Combinatorial Chemistry Techniques , Furans/chemistry , HIV Reverse Transcriptase/chemistry , Humans , Reverse Transcriptase Inhibitors/chemistry , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Thiophenes/chemistry
20.
J Biol Chem ; 283(14): 9196-205, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18218634

ABSTRACT

The 2',3'-dideoxy-3'-thiacytidine drug-resistant M184I HIV-1 reverse transcriptase (RT) has been shown to synthesize DNA with decreased processivity compared with the wild-type RT. M184A displays an even more severe processivity defect. However, the basis of this decreased processivity has been unclear, and both primer-template binding and dNTP interaction defects have been proposed to account for it. In this study, we show that the altered properties of the M184I and M184A RT mutants that we have measured, including decreased processivity, a slower rate of primer extension, and increased strand transfer activity, can all be explained by a defect in dNTP utilization. These alterations are observed only at low dNTP concentration and vanish as the dNTP concentration is raised. The mutant RTs exhibit a normal dissociation rate from a DNA primer-RNA template while paused during synthesis. Slower than normal synthesis at physiological dNTP concentration, coupled with normal dissociation from the primer-template, results in the lowered processivity. The mutant RTs exhibit normal DNA 3'-end-directed and RNA 5'-end-directed ribonuclease H activity. The reduced rate of DNA synthesis causes an increase in the ratio of ribonuclease H to polymerase activity thereby promoting increased strand transfer. These latter results are consistent with an observed higher rate of recombination by HIV-1 strains with Met-184 mutations.


Subject(s)
DNA Primers/chemistry , DNA, Viral/chemistry , Deoxyribonucleotides/chemistry , HIV Reverse Transcriptase/chemistry , HIV-1/enzymology , Mutation, Missense , Amino Acid Substitution , DNA Primers/genetics , DNA Primers/metabolism , DNA, Viral/biosynthesis , DNA, Viral/genetics , Deoxyribonucleotides/metabolism , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , Lamivudine/chemistry , Lamivudine/pharmacology , Recombination, Genetic/drug effects , Recombination, Genetic/genetics , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...