Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Med Chem ; 54(8): 2727-37, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21446702

ABSTRACT

tert-Butyldimethylsilyl-spiroaminooxathioledioxide (TSAO) compounds have an embedded thymidine-analogue backbone; however, TSAO compounds invoke non-nucleoside RT inhibitor (NNRTI) resistance mutations. Our crystal structure of RT:7 (TSAO-T) complex shows that 7 binds inside the NNRTI-binding pocket, assuming a "dragon" shape, and interacts extensively with almost all the pocket residues. The structure also explains the structure-activity relationships and resistance data for TSAO compounds. The binding of 7 causes hyper-expansion of the pocket and significant rearrangement of RT subdomains. This nonoptimal complex formation is apparently responsible (1) for the lower stability of a RT (p66/p51) dimer and (2) for the lower potency of 7 despite of its extensive interactions with RT. However, the HIV-1 RT:7 structure reveals novel design features such as (1) interactions with the conserved Tyr183 from the YMDD-motif and (2) a possible way for an NNRTI to reach the polymerase active site that may be exploited in designing new NNRTIs.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Spiro Compounds/chemistry , Thymidine/analogs & derivatives , Crystallography, X-Ray , Elasticity , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Thymidine/chemistry , Uridine/analogs & derivatives
2.
Nat Struct Mol Biol ; 17(10): 1202-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20852643

ABSTRACT

Human immunodeficiency virus (HIV-1) develops resistance to 3'-azido-2',3'-deoxythymidine (AZT, zidovudine) by acquiring mutations in reverse transcriptase that enhance the ATP-mediated excision of AZT monophosphate from the 3' end of the primer. The excision reaction occurs at the dNTP-binding site, uses ATP as a pyrophosphate donor, unblocks the primer terminus and allows reverse transcriptase to continue viral DNA synthesis. The excision product is AZT adenosine dinucleoside tetraphosphate (AZTppppA). We determined five crystal structures: wild-type reverse transcriptase-double-stranded DNA (RT-dsDNA)-AZTppppA; AZT-resistant (AZTr; M41L D67N K70R T215Y K219Q) RT-dsDNA-AZTppppA; AZTr RT-dsDNA terminated with AZT at dNTP- and primer-binding sites; and AZTr apo reverse transcriptase. The AMP part of AZTppppA bound differently to wild-type and AZTr reverse transcriptases, whereas the AZT triphosphate part bound the two enzymes similarly. Thus, the resistance mutations create a high-affinity ATP-binding site. The structure of the site provides an opportunity to design inhibitors of AZT-monophosphate excision.


Subject(s)
Drug Resistance, Viral/physiology , HIV Reverse Transcriptase/chemistry , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Zidovudine/pharmacology , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Binding Sites/drug effects , Crystallography, X-Ray , DNA, Viral/biosynthesis , Deoxyribonucleotides/metabolism , Dideoxynucleotides/metabolism , Drug Design , Drug Resistance, Viral/genetics , Genes, rev , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , HIV-1/genetics , Models, Molecular , Mutation, Missense , Point Mutation , Protein Conformation , Structure-Activity Relationship , Thymine Nucleotides/metabolism , Zidovudine/analogs & derivatives , Zidovudine/metabolism
3.
Structure ; 17(12): 1625-1635, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-20004166

ABSTRACT

Novel inhibitors are needed to counteract the rapid emergence of drug-resistant HIV variants. HIV-1 reverse transcriptase (RT) has both DNA polymerase and RNase H (RNH) enzymatic activities, but approved drugs that inhibit RT target the polymerase. Inhibitors that act against new targets, such as RNH, should be effective against all of the current drug-resistant variants. Here, we present 2.80 A and 2.04 A resolution crystal structures of an RNH inhibitor, beta-thujaplicinol, bound at the RNH active site of both HIV-1 RT and an isolated RNH domain. beta-thujaplicinol chelates two divalent metal ions at the RNH active site. We provide biochemical evidence that beta-thujaplicinol is a slow-binding RNH inhibitor with noncompetitive kinetics and suggest that it forms a tropylium ion that interacts favorably with RT and the RNA:DNA substrate.


Subject(s)
HIV Reverse Transcriptase/chemistry , Reverse Transcriptase Inhibitors/chemistry , Ribonucleases/metabolism , Tropolone/analogs & derivatives , Catalytic Domain , Crystallography, X-Ray , HIV Reverse Transcriptase/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Reverse Transcriptase Inhibitors/metabolism , Tropolone/chemistry , Tropolone/metabolism
4.
J Biol Chem ; 284(50): 35092-100, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19812032

ABSTRACT

K65R is a primary reverse transcriptase (RT) mutation selected in human immunodeficiency virus type 1-infected patients taking antiretroviral regimens containing tenofovir disoproxil fumarate or other nucleoside analog RT drugs. We determined the crystal structures of K65R mutant RT cross-linked to double-stranded DNA and in complexes with tenofovir diphosphate (TFV-DP) or dATP. The crystals permit substitution of TFV-DP with dATP at the dNTP-binding site. The guanidinium planes of the arginines K65R and Arg(72) were stacked to form a molecular platform that restricts the conformational adaptability of both of the residues, which explains the negative effects of the K65R mutation on nucleotide incorporation and on excision. Furthermore, the guanidinium planes of K65R and Arg(72) were stacked in two different rotameric conformations in TFV-DP- and dATP-bound structures that may help explain how K65R RT discriminates the drug from substrates. These K65R-mediated effects on RT structure and function help us to visualize the complex interaction with other key nucleotide RT drug resistance mutations, such as M184V, L74V, and thymidine analog resistance mutations.


Subject(s)
Adenine/analogs & derivatives , Drug Resistance, Viral/physiology , HIV Reverse Transcriptase , Mutation , Organophosphonates/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Adenine/chemistry , Adenine/pharmacology , Arginine/genetics , Arginine/metabolism , Crystallization , Crystallography, X-Ray , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/drug effects , HIV Reverse Transcriptase/physiology , Humans , Models, Molecular , Molecular Sequence Data , Molecular Structure , Organophosphonates/chemistry , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry , Tenofovir
5.
Nucleic Acids Res ; 36(15): 5083-92, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18676450

ABSTRACT

HIV-1 reverse transcriptase (RT) is a primary target for anti-AIDS drugs. Structures of HIV-1 RT, usually determined at approximately 2.5-3.0 A resolution, are important for understanding enzyme function and mechanisms of drug resistance in addition to being helpful in the design of RT inhibitors. Despite hundreds of attempts, it was not possible to obtain the structure of a complex of HIV-1 RT with TMC278, a nonnucleoside RT inhibitor (NNRTI) in advanced clinical trials. A systematic and iterative protein crystal engineering approach was developed to optimize RT for obtaining crystals in complexes with TMC278 and other NNRTIs that diffract X-rays to 1.8 A resolution. Another form of engineered RT was optimized to produce a high-resolution apo-RT crystal form, reported here at 1.85 A resolution, with a distinct RT conformation. Engineered RTs were mutagenized using a new, flexible and cost effective method called methylated overlap-extension ligation independent cloning. Our analysis suggests that reducing the solvent content, increasing lattice contacts, and stabilizing the internal low-energy conformations of RT are critical for the growth of crystals that diffract to high resolution. The new RTs enable rapid crystallization and yield high-resolution structures that are useful in designing/developing new anti-AIDS drugs.


Subject(s)
Crystallography, X-Ray , HIV Reverse Transcriptase/chemistry , Nitriles/chemistry , Protein Engineering/methods , Pyrimidines/chemistry , Reverse Transcriptase Inhibitors/chemistry , Cloning, Molecular , Drug Design , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Models, Molecular , Mutagenesis , Rilpivirine
6.
Proc Natl Acad Sci U S A ; 105(5): 1466-71, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18230722

ABSTRACT

TMC278 is a diarylpyrimidine (DAPY) nonnucleoside reverse transcriptase inhibitor (NNRTI) that is highly effective in treating wild-type and drug-resistant HIV-1 infections in clinical trials at relatively low doses ( approximately 25-75 mg/day). We have determined the structure of wild-type HIV-1 RT complexed with TMC278 at 1.8 A resolution, using an RT crystal form engineered by systematic RT mutagenesis. This high-resolution structure reveals that the cyanovinyl group of TMC278 is positioned in a hydrophobic tunnel connecting the NNRTI-binding pocket to the nucleic acid-binding cleft. The crystal structures of TMC278 in complexes with the double mutant K103N/Y181C (2.1 A) and L100I/K103N HIV-1 RTs (2.9 A) demonstrated that TMC278 adapts to bind mutant RTs. In the K103N/Y181C RT/TMC278 structure, loss of the aromatic ring interaction caused by the Y181C mutation is counterbalanced by interactions between the cyanovinyl group of TMC278 and the aromatic side chain of Y183, which is facilitated by an approximately 1.5 A shift of the conserved Y(183)MDD motif. In the L100I/K103N RT/TMC278 structure, the binding mode of TMC278 is significantly altered so that the drug conforms to changes in the binding pocket primarily caused by the L100I mutation. The flexible binding pocket acts as a molecular "shrink wrap" that makes a shape complementary to the optimized TMC278 in wild-type and drug-resistant forms of HIV-1 RT. The crystal structures provide a better understanding of how the flexibility of an inhibitor can compensate for drug-resistance mutations.


Subject(s)
Anti-HIV Agents/chemistry , Drug Resistance, Viral/genetics , Enzyme Inhibitors/chemistry , HIV Reverse Transcriptase/chemistry , Nitriles/chemistry , Pyrimidines/chemistry , Reverse Transcriptase Inhibitors/chemistry , Amino Acid Motifs/genetics , Amino Acid Sequence , Crystallography, X-Ray , Drug Design , HIV Reverse Transcriptase/genetics , Humans , Molecular Sequence Data , Mutation , Protein Conformation , Protein Engineering , Rilpivirine
7.
J Mol Biol ; 365(1): 77-89, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17056061

ABSTRACT

Lys103Asn and Tyr181Cys are the two mutations frequently observed in patients exposed to various non-nucleoside reverse transcriptase inhibitor drugs (NNRTIs). Human immunodeficiency virus (HIV) strains containing both reverse transcriptase (RT) mutations are resistant to all of the approved NNRTI drugs. We have determined crystal structures of Lys103Asn/Tyr181Cys mutant HIV-1 RT with and without a bound non-nucleoside inhibitor (HBY 097, (S)-4-isopropoxycarbonyl-6-methoxy-3-(methylthio-methyl)-3,4-dihydroquinoxalin-2(1H)-thione) at 3.0 A and 2.5 A resolution, respectively. The structure of the double mutant RT/HBY 097 complex shows a rearrangement of the isopropoxycarbonyl group of HBY 097 compared to its binding with wild-type RT. HBY 097 makes a hydrogen bond with the thiol group of Cys181 that helps the drug retain potency against the Tyr181Cys mutation. The structure of the unliganded double mutant HIV-1 RT showed that Lys103Asn mutation facilitates coordination of a sodium ion with Lys101 O, Asn103 N and O(delta1), Tyr188 O(eta), and two water molecules. The formation of the binding pocket requires the removal of the sodium ion. Although the RT alone and the RT/HBY 097 complex were crystallized in the presence of ATP, only the RT has an ATP coordinated with two Mn(2+) at the polymerase active site. The metal coordination mimics a reaction intermediate state in which complete octahedral coordination was observed for both metal ions. Asp186 coordinates at an axial position whereas the carboxylates of Asp110 and Asp185 are in the planes of coordination of both metal ions. The structures provide evidence that NNRTIs restrict the flexibility of the YMDD loop and prevent the catalytic aspartate residues from adopting their metal-binding conformations.


Subject(s)
Adenosine Triphosphate/metabolism , Anti-HIV Agents/metabolism , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/enzymology , Amino Acid Substitution , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Binding Sites , Crystallography, X-Ray , DNA/metabolism , Drug Resistance, Viral , HIV Reverse Transcriptase/antagonists & inhibitors , Humans , Manganese/metabolism , Models, Molecular , Molecular Conformation , Mutation , Protein Conformation , Quinoxalines/chemistry , Quinoxalines/metabolism , Quinoxalines/pharmacology , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/pharmacology
8.
ACS Chem Biol ; 1(11): 702-12, 2006 Dec 20.
Article in English | MEDLINE | ID: mdl-17184135

ABSTRACT

The rapid emergence of drug-resistant variants of human immunodeficiency virus, type 1 (HIV-1), has limited the efficacy of anti-acquired immune deficiency syndrome (AIDS) treatments, and new lead compounds that target novel binding sites are needed. We have determined the 3.15 A resolution crystal structure of HIV-1 reverse transcriptase (RT) complexed with dihydroxy benzoyl naphthyl hydrazone (DHBNH), an HIV-1 RT RNase H (RNH) inhibitor (RNHI). DHBNH is effective against a variety of drug-resistant HIV-1 RT mutants. While DHBNH has little effect on most aspects of RT-catalyzed DNA synthesis, at relatively high concentrations it does inhibit the initiation of RNA-primed DNA synthesis. Although primarily an RNHI, DHBNH binds >50 A away from the RNH active site, at a novel site near both the polymerase active site and the non-nucleoside RT inhibitor (NNRTI) binding pocket. When DHBNH binds, both Tyr181 and Tyr188 remain in the conformations seen in unliganded HIV-1 RT. DHBNH interacts with conserved residues (Asp186, Trp229) and has substantial interactions with the backbones of several less well-conserved residues. On the basis of this structure, we designed substituted DHBNH derivatives that interact with the NNRTI-binding pocket. These compounds inhibit both the polymerase and RNH activities of RT.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/chemistry , Reverse Transcriptase Inhibitors/chemistry , Ribonuclease H/antagonists & inhibitors , Cell Line, Tumor , HIV Reverse Transcriptase/metabolism , Humans , Hydrazones/chemistry , Hydrazones/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary/drug effects , Protein Structure, Secondary/physiology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H/metabolism , Structure-Activity Relationship
9.
J Med Chem ; 48(24): 7582-91, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16302798

ABSTRACT

In the treatment of AIDS, the efficacy of all drugs, including non-nucleoside inhibitors (NNRTIs) of HIV-1 reverse transcriptase (RT), has been limited by the rapid appearance of drug-resistant viruses. Lys103Asn, Tyr181Cys, and Tyr188Leu are some of the most common RT mutations that cause resistance to NNRTIs in the clinic. We report X-ray crystal structures for RT complexed with three different pyridinone derivatives, R157208, R165481, and R221239, at 2.95, 2.9, and 2.43 A resolution, respectively. All three ligands exhibit nanomolar or subnanomolar inhibitory activity against wild-type RT, but varying activities against drug-resistant mutants. R165481 and R221239 differ from most NNRTIs in that binding does not involve significant contacts with Tyr181. These compounds strongly inhibit wild-type HIV-1 RT and drug-resistant variants, including Tyr181Cys and Lys103Asn RT. These properties result in part from an iodine atom on the pyridinone ring of both inhibitors that interacts with the main-chain carbonyl oxygen of Tyr188. An acrylonitrile substituent on R165481 substantially improves the activity of the compound against wild-type RT (and several mutants) and provides a way to generate novel inhibitors that could interact with conserved elements of HIV-1 RT at the polymerase catalytic site. In R221239, there is a flexible linker to a furan ring that permits interactions with Val106, Phe227, and Pro236. These contacts appear to enhance the inhibitory activity of R221239 against the HIV-1 strains that carry the Val106Ala, Tyr188Leu, and Phe227Cys mutations.


Subject(s)
Drug Resistance, Viral , HIV Reverse Transcriptase/chemistry , HIV-1/enzymology , Pyridones/chemistry , Reverse Transcriptase Inhibitors/chemistry , Crystallography, X-Ray , HIV-1/genetics , Models, Molecular , Molecular Structure , Mutation
10.
Cell ; 122(4): 541-52, 2005 Aug 26.
Article in English | MEDLINE | ID: mdl-16122422

ABSTRACT

We define the target, mechanism, and structural basis of inhibition of bacterial RNA polymerase (RNAP) by the tetramic acid antibiotic streptolydigin (Stl). Stl binds to a site adjacent to but not overlapping the RNAP active center and stabilizes an RNAP-active-center conformational state with a straight-bridge helix. The results provide direct support for the proposals that alternative straight-bridge-helix and bent-bridge-helix RNAP-active-center conformations exist and that cycling between straight-bridge-helix and bent-bridge-helix RNAP-active-center conformations is required for RNAP function. The results set bounds on models for RNAP function and suggest strategies for design of novel antibacterial agents.


Subject(s)
Aminoglycosides/pharmacology , Bacteria/enzymology , Bacteria/genetics , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , RNA, Messenger/biosynthesis , Aminoglycosides/chemistry , Binding Sites/drug effects , Binding Sites/physiology , DNA-Directed RNA Polymerases/chemistry , Feedback, Physiological/physiology , Models, Molecular , Molecular Structure , Protein Structure, Secondary/drug effects , Protein Structure, Secondary/genetics
11.
J Med Chem ; 48(6): 1974-83, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15771441

ABSTRACT

We have examined selected physicochemical properties of compounds from the diaryltriazine/diarylpyrimidine (DATA/DAPY) classes of non-nucleoside reverse transcriptase inhibitors (NNRTIs) and explored possible correlations with their bioavailability. In simple aqueous solutions designed to mimic the gastrointestinal (GI) environment of a fasting individual, all NNRTIs demonstrated formation of aggregates as detected by dynamic light scattering and electron microscopy. Under various conditions mimicking physiological transitions in the GI environment, aggregate size distributions were shown to depend on compound concentration and pH. NNRTIs with good absorption were capable of forming aggregates with hydrodynamic radii of /=250 nm at concentrations above 0.01 mM, probably representing precipitate. We propose a model in which the uptake rate into systemic circulation depends on having hydrophobic drug aggregates of appropriate size available for absorption at different locations within the GI tract.


Subject(s)
Biological Availability , Models, Biological , Pyrimidines/chemistry , Reverse Transcriptase Inhibitors/chemistry , Triazines/chemistry , Absorption , Administration, Oral , Animals , Chemical Phenomena , Chemistry, Physical , Gastrointestinal Tract , Half-Life , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Light , Microscopy, Electron, Transmission , Rats , Scattering, Radiation , Solutions
12.
J Med Chem ; 47(10): 2550-60, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15115397

ABSTRACT

Anti-AIDS drug candidate and non-nucleoside reverse transcriptase inhibitor (NNRTI) TMC125-R165335 (etravirine) caused an initial drop in viral load similar to that observed with a five-drug combination in naïve patients and retains potency in patients infected with NNRTI-resistant HIV-1 variants. TMC125-R165335 and related anti-AIDS drug candidates can bind the enzyme RT in multiple conformations and thereby escape the effects of drug-resistance mutations. Structural studies showed that this inhibitor and other diarylpyrimidine (DAPY) analogues can adapt to changes in the NNRTI-binding pocket in several ways: (1). DAPY analogues can bind in at least two conformationally distinct modes; (2). within a given binding mode, torsional flexibility ("wiggling") of DAPY analogues permits access to numerous conformational variants; and (3). the compact design of the DAPY analogues permits significant repositioning and reorientation (translation and rotation) within the pocket ("jiggling"). Such adaptations appear to be critical for potency against wild-type and a wide range of drug-resistant mutant HIV-1 RTs. Exploitation of favorable components of inhibitor conformational flexibility (such as torsional flexibility about strategically located chemical bonds) can be a powerful drug design concept, especially for designing drugs that will be effective against rapidly mutating targets.


Subject(s)
Anti-HIV Agents/chemistry , Drug Resistance, Viral , HIV Reverse Transcriptase/chemistry , Pyridazines/chemistry , Reverse Transcriptase Inhibitors/chemistry , Crystallography, X-Ray , HIV Reverse Transcriptase/genetics , Models, Molecular , Mutation , Nitriles , Protein Conformation , Pyrimidines/chemistry
13.
Nat Struct Mol Biol ; 11(5): 469-74, 2004 May.
Article in English | MEDLINE | ID: mdl-15107837

ABSTRACT

Tenofovir, also known as PMPA, R-9-(2-(phosphonomethoxypropyl)adenine, is a nucleotide reverse transcriptase (RT) inhibitor. We have determined the crystal structures of two related complexes of HIV-1 RT with template primer and tenofovir: (i) a ternary complex at a resolution of 3.0 A of RT crosslinked to a dideoxy-terminated DNA with tenofovir-diphosphate bound as the incoming substrate; and (ii) a RT-DNA complex at a resolution of 3.1 A with tenofovir at the 3' primer terminus. The tenofovir nucleotide in the tenofovir-terminated structure seems to adopt multiple conformations. Some nucleoside reverse transcriptase inhibitors, including 3TC and AZT, have elements ('handles') that project beyond the corresponding elements on normal dNTPs (the 'substrate envelope'). HIV-1 RT resistance mechanisms to AZT and 3TC take advantage of these handles; tenofovir's structure lacks handles that could protrude through the substrate envelope to cause resistance.


Subject(s)
Adenine/analogs & derivatives , Adenine/chemistry , DNA, Viral/chemistry , HIV Reverse Transcriptase/chemistry , Organophosphonates , Organophosphorus Compounds/chemistry , Reverse Transcriptase Inhibitors/chemistry , Base Sequence , DNA Primers , Models, Molecular , Tenofovir
14.
Structure ; 12(4): 657-67, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15062088

ABSTRACT

Arginine deiminase (ADI), an enzyme that hydrolyzes arginine to generate energy in many parasitic microorganisms, has potent anticancer activities and can halt growth of solid tumors. We determined the crystal structure of ADI from Mycoplasma arginini in two different forms (1.6 and 2.0 A resolution) using multiple isomorphous replacement. ADI shares common structural features with the arginine-catabolizing enzymes Arg:Gly amidinotransferase and dimethylarginine dimethyl-aminohydrolase; ADI contains an additional domain of five helices. The scissile C-N bonds of the substrates and the catalytic triads (Cys398-His269-Glu213 of ADI) for the three enzymes superimpose on each other. The ADI structure from form I crystals corresponds to a tetrahedral intermediate with four heteroatoms (1S, 2N, 1O) covalently bonded to the reaction-center carbon. The structure from form II crystals represents an amidino-enzyme complex; the reaction-center carbon is covalently bonded to Cys398 sulfur and two nitrogens, and the reacting water molecule is only 2.54 A away.


Subject(s)
Hydrolases/chemistry , Amino Acid Sequence , Arginine/metabolism , Citrulline/metabolism , Crystallography, X-Ray , Hydrolases/isolation & purification , Hydrolases/metabolism , Molecular Sequence Data , Mycoplasma/enzymology , Protein Structure, Tertiary
15.
J Biol Chem ; 278(18): 16280-8, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12554739

ABSTRACT

A disulfide cross-linking strategy was used to covalently trap as a stable complex (complex N) a short-lived, kinetic intermediate in DNA polymerization. This intermediate corresponds to the product of polymerization prior to translocation. We also prepared the trapped complex that corresponds to the product of polymerization after translocation (complex P). The cross-linking method that we used is a variation of a technique developed by the Verdine and Harrison laboratories. It involves disulfide interchange between an engineered sulfhydryl group of the protein (Q258C mutation) and a disulfide-containing tether attached at the N(2) amino group of a modified dG in either the template or the primer strand of the nucleic acid. We report here a highly efficient synthesis of the precursor, bis(3-aminopropyl)disulfide dihydrochloride, used to introduce this substituent into the oligonucleotide. Efficient cross-linking takes place when the base pair containing the substituent is positioned seven registers from the dNTP-binding site (N site) and the N site is occupied. Complex N, but not complex P, is a substrate for the ATP-based excision reaction that unblocks nucleoside reverse transcriptase inhibitor (NRTI)-terminated primers and causes resistance to several NRTIs, confirming predictions that the excision reaction takes place only when the 3'-end of the primer is bound at the N site. These techniques can be used for biochemical and structural studies of the mechanism of DNA polymerization, translocation, and excision-based resistance of RT to NRTIs. They may also be useful in studying other DNA or RNA polymerases or other enzymes.


Subject(s)
DNA/metabolism , HIV Reverse Transcriptase/metabolism , Adenosine Triphosphate/metabolism , Biological Transport , Crystallization , HIV Reverse Transcriptase/chemistry
16.
EMBO J ; 21(23): 6614-24, 2002 Dec 02.
Article in English | MEDLINE | ID: mdl-12456667

ABSTRACT

AZT (3'-azido-3'-deoxythymidine) resistance involves the enhanced excision of AZTMP from the end of the primer strand by HIV-1 reverse transcriptase. This reaction can occur when an AZTMP-terminated primer is bound at the nucleotide-binding site (pre-translocation complex N) but not at the 'priming' site (post-translocation complex P). We determined the crystal structures of N and P complexes at 3.0 and 3.1 A resolution. These structures provide insight into the structural basis of AZTMP excision and the mechanism of translocation. Docking of a dNTP in the P complex structure suggests steric crowding in forming a stable ternary complex that should increase the relative amount of the N complex, which is the substrate for excision. Structural differences between complexes N and P suggest that the conserved YMDD loop is involved in translocation, acting as a springboard that helps to propel the primer terminus from the N to the P site after dNMP incorporation.


Subject(s)
DNA/metabolism , HIV-1/metabolism , RNA-Directed DNA Polymerase/metabolism , Thymine Nucleotides/metabolism , Zidovudine/analogs & derivatives , Zidovudine/metabolism , DNA/biosynthesis , Dideoxynucleotides , Drug Resistance, Viral/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...