Journal of Molecular Biology
Volume 385, Issue 2, 16 January 2009, Pages 568-579
Journal home page for Journal of Molecular Biology

Defining the DNA Substrate Binding Sites on HIV-1 Integrase

https://doi.org/10.1016/j.jmb.2008.10.083Get rights and content

Abstract

A tetramer model for human immunodeficiency virus type 1 (HIV-1) integrase (IN) with DNA representing long terminal repeat (LTR) termini was previously assembled to predict the IN residues that interact with the LTR termini; these predictions were experimentally verified for nine amino acid residues [Chen, A., Weber, I. T., Harrison, R. W. & Leis, J. (2006). Identification of amino acids in HIV-1 and avian sarcoma virus integrase subsites required for specific recognition of the long terminal repeat ends. J. Biol. Chem., 281, 4173–4182]. In a similar strategy, the unique amino acids found in avian sarcoma virus IN, rather than HIV-1 or Mason–Pfizer monkey virus IN, were substituted into the structurally related positions of HIV-1 IN. Substitutions of six additional residues (Q44, L68, E69, D229, S230, and D253) showed changes in the 3′ processing specificity of the enzyme, verifying their predicted interaction with the LTR DNA. The newly identified residues extend interactions along a 16-bp length of the LTR termini and are consistent with known LTR DNA/HIV-1 IN cross-links. The tetramer model for HIV-1 IN with LTR termini was modified to include two IN binding domains for lens-epithelium-derived growth factor/p75. The target DNA was predicted to bind in a surface trench perpendicular to the plane of the LTR DNA binding sites of HIV-1 IN and extending alongside lens-epithelium-derived growth factor. This hypothesis is supported by the in vitro activity phenotype of HIV-1 IN mutant, with a K219S substitution showing loss in strand transfer activity while maintaining 3′ processing on an HIV-1 substrate. Mutations at seven other residues reported in the literature have the same phenotype, and all eight residues align along the length of the putative target DNA binding trench.

Introduction

The integrase (IN) of human immunodeficiency virus type 1 (HIV-1) is an attractive target for therapeutic development, as it is essential for early steps in viral replication and there are no homologues in the eukaryotic system for which inhibitors would negatively affect host viability. This enzyme is both necessary and sufficient to catalyze the insertion of viruses into host DNA.1, 2, 3 In the first step or 3′ processing reaction, two deoxyribonucleotides are removed from the 3′ end of long terminal repeat (LTR) strands containing the highly conserved CA dinucleotides. In the second step or strand transfer reaction, the newly created 3′ ends undergo a staggered nucleophilic attack on two strands of the target DNA. These structures are resolved and repaired by host cell enzymes, resulting in an integrated copy of the viral DNA, with the gene-encoding sequence colinear to the viral RNA and flanked by a 4- to 6-bp duplication of the target DNA, depending upon the viral IN.

Design of inhibitors has been hampered by the lack of crystal structures available for full-length IN monomers or higher-order IN oligomers, let alone in complex with DNA. Partial structures with two of the three domains have been reported, and these were used to assemble a model of a tetramer HIV-1 IN with bound LTR DNAs.4 The model was then used to predict residues that were in close proximity to the viral DNA. To verify these predictions, a structural alignment of the primary sequences of HIV-1, simian immunodeficiency virus (SIV), and avian sarcoma virus (ASV) INs5 was used to identify residues that were unique to each virus enzyme, since viral IN specifically recognizes its cognate LTR end substrates. The unique amino acids from ASV IN were substituted into the equivalent structural position of HIV-1 IN. Substitution of the ASV IN residues conferred on the HIV-1 IN mutants the partial ability to cleave an ASV substrate. Multiple residues of HIV-1 IN were demonstrated to alter specificity (V72, S153, K160, I161, G163, Q164, V165, H171, and L172,4 shown in red in Fig. 1) for only one of the two LTR ends. In this report, we have identified six additional HIV-1 IN residues that influence the selection of the LTR end substrates for 3′ processing. These include Q44, L68, E69, D229, S230, and D253, which align, along the two LTR binding grooves, with previous residues that alter recognition.

In addition, the structural model was modified to include the host protein lens-epithelium-derived growth factor (LEDGF)/p75 IN binding domains and to predict a trench on the HIV-1 IN surface that may accommodate the target DNA. The putative binding site for target DNA is positioned roughly perpendicular to the LTR binding sites. Consistent with this interpretation, we have identified in the literature a series of amino acids along the length of one side of this trench, where point amino acid substitutions result in enzymes that lose the ability to strand transfer with little or no effect on 3′ processing. In addition, we have identified a residue on the opposite wall of the trench, K219, where serine substitution displays the same phenotype. This residue was found in a peptide that was previously demonstrated to cross-link to the target DNA.7

Section snippets

Prediction of additional HIV-1 IN residues interacting with LTR DNA ends

In the original selection of amino acids that could affect the recognition of LTR ends, we used the structural alignment of SIV, HIV-1, and ASV INs to identify those residues that were unique. We subsequently observed that HIV-1 IN was capable of 3′ processing a U5 SIV, but not a Mason–Pfizer monkey virus, LTR DNA substrate (data not shown). Therefore, we examined the structural alignment of HIV-1 and ASV INs with the Mason–Pfizer monkey virus IN sequence.5 This analysis identified additional

Discussion

The homotetramer form of IN catalyzes all of its known enzymatic activities. While dimers of IN are capable of catalyzing 3′ processing and strand transfer reactions, they do not support a concerted DNA integration reaction.27 For this reason, a homotetramer model was assembled. In the model, two of the four subunits are depicted with major contacts with the LTR and target DNAs. The remaining two subunits are available for contacts with proteins that interact with IN,28, 29 including the host

Reagents

[γ-33P]ATP (2500 Ci/mmol) was purchased from Perkin Elmer Life Sciences. HiTrap™ Chelating HP resin and HiTrap™ Heparin HP resin were purchased from GE Healthcare Life Sciences (Piscataway, NJ). T4 polynucleotide kinase was obtained from USB (Cleveland, OH). IPTG was obtained from Roche (Indianapolis, IN). The Slide-A-Lyzer Dialysis cassette (molecular weight cutoff, 10,000) was obtained from Pierce (Rockford, IL). CentriPrep centrifugal filter devices with YM-10 MW membranes were obtained from

Acknowledgements

We thank Yuan-Fang Wang for assistance with modeling and preparation of structural figures. This work was supported, in part, by United States Public Health Service grants AI054143 (to J.L.), GM6290, and GM065762 (to I.T.W. and R.W.H.). J.D. was supported, in part, by the Training Program in Viral Replication T32 AI060523.

References (71)

  • Van MaeleB. et al.

    Cellular co-factors of HIV-1 integration

    Trends Biochem. Sci.

    (2006)
  • CherepanovP. et al.

    HIV-1 integrase forms stable tetramers and associates with LEDGF/p75 protein in human cells

    J. Biol. Chem.

    (2003)
  • JenkinsT.M. et al.

    A soluble active mutant of HIV-1 integrase: involvement of both the core and carboxyl-terminal domains in multimerization

    J. Biol. Chem.

    (1996)
  • AgapkinaJ. et al.

    Probing of HIV-1 integrase/DNA interactions using novel analogs of viral DNA

    J. Biol. Chem.

    (2006)
  • GuareJ.P. et al.

    A series of 5-aminosubstituted 4-fluorobenzyl-8-hydroxy-[1,6]naphthyridine-7-carboxamide HIV-1 integrase inhibitors

    Bioorg. Med. Chem. Lett.

    (2006)
  • EmbreyM.W. et al.

    A series of 5-(5,6)-dihydrouracil substituted 8-hydroxy-[1,6]naphthyridine-7-carboxylic acid 4-fluorobenzylamide inhibitors of HIV-1 integrase and viral replication in cells

    Bioorg. Med. Chem. Lett.

    (2005)
  • MarchandC. et al.

    Structural determinants for HIV-1 integrase inhibition by beta-diketo acids

    J. Biol. Chem.

    (2002)
  • MaertensG. et al.

    LEDGF/p75 is essential for nuclear and chromosomal targeting of HIV-1 integrase in human cells

    J. Biol. Chem.

    (2003)
  • EmilianiS. et al.

    Integrase mutants defective for interaction with LEDGF/p75 are impaired in chromosome tethering and HIV-1 replication

    J. Biol. Chem.

    (2005)
  • JeltschA. et al.

    Engineering novel restriction endonucleases: principles and applications

    Trends Biotechnol.

    (1996)
  • JohnsonA.A. et al.

    Integration requires a specific interaction of the donor DNA terminal 5′-cytosine with glutamine 148 of the HIV-1 integrase flexible loop

    J. Biol. Chem.

    (2006)
  • PrietS. et al.

    Reversion of the lethal phenotype of an HIV-1 integrase mutant virus by overexpression of the same integrase mutant protein

    J. Biol. Chem.

    (2003)
  • SayasithK. et al.

    Characterization of mutant HIV-1 integrase carrying amino acid changes in the catalytic domain

    Mol. Cells

    (2000)
  • BrinE. et al.

    Changes in the mechanism of DNA integration in vitro induced by base substitutions in the HIV-1 U5 and U3 terminal sequences

    J. Biol. Chem.

    (2002)
  • BrinE. et al.

    HIV-1 integrase interaction with U3 and U5 terminal sequences in vitro defined using substrates with random sequences

    J. Biol. Chem.

    (2002)
  • SinhaS. et al.

    Recombinant human immunodeficiency virus type 1 integrase exhibits a capacity for full-site integration in vitro that is comparable to that of purified preintegration complexes from virus-infected cells

    J. Virol.

    (2005)
  • SnaselJ. et al.

    Integrase of Mason–Pfizer monkey virus

    FEBS J.

    (2005)
  • De LanoW.L.

    The PyMol Molecular Graphics System v1.00 edit

    (2002)
  • HeuerT.S. et al.

    Mapping features of HIV-1 integrase near selected sites on viral and target DNA molecules in an active enzyme–DNA complex by photo-cross-linking

    Biochemistry

    (1997)
  • BischerourJ. et al.

    The (52–96) C-terminal domain of Vpr stimulates HIV-1 IN-mediated homologous strand transfer of mini-viral DNA

    Nucleic Acids Res.

    (2003)
  • EngelmanA. et al.

    Structure-based mutagenesis of the catalytic domain of human immunodeficiency virus type 1 integrase

    J. Virol.

    (1997)
  • ZhuK. et al.

    Requirement for integrase during reverse transcription of human immunodeficiency virus type 1 and the effect of cysteine mutations of integrase on its interactions with reverse transcriptase

    J. Virol.

    (2004)
  • FikkertV. et al.

    Development of resistance against diketo derivatives of human immunodeficiency virus type 1 by progressive accumulation of integrase mutations

    J. Virol.

    (2003)
  • HazudaD.J. et al.

    A naphthyridine carboxamide provides evidence for discordant resistance between mechanistically identical inhibitors of HIV-1 integrase

    Proc. Natl Acad. Sci. USA

    (2004)
  • CherepanovP. et al.

    Structural basis for the recognition between HIV-1 integrase and transcriptional coactivator p75

    Proc. Natl Acad. Sci. USA

    (2005)
  • Cited by (26)

    • New insights into the interaction between pyrrolyl diketoacids and HIV-1 integrase active site and comparison with RNase H

      2016, Antiviral Research
      Citation Excerpt :

      Such residues play important structural roles since they belong to the 140–149 loop of the catalytic-core domain, which is required for efficient IN function (Mouscadet et al., 2010). In particular, P145 amino acid residue was reported to establish a hydrogen bond with Q148, central to maintain the 140–149 loop in a conformation favorable to RAL binding (Chen et al., 2013), while Y143, Q146 and Q148 were predicted to constitute part of the DNA substrate binding site (Dolan et al., 2009). To have a complete overview of the behavior of pyrrolyl DKAs in presence of the three patterns involved in resistance to INIs, mutagenesis studies were also performed on the N155 amino acid residue, since the N155H mutation has been reported to disrupt the molecular environment needed for RAL optimal binding through the displacement of the DNA backbone and the catalytic DDE carboxylates (Hare et al., 2010a).

    • Structural properties of HIV integrase·lens epithelium-derived growth factor oligomers

      2010, Journal of Biological Chemistry
      Citation Excerpt :

      There are currently no crystal structures available containing full-length HIV IN. Thus, the intermolecular contacts observed in two-domain crystal structures have been used to construct plausible models of higher oligomeric forms of the IN protein (18, 25, 26, 34, 35, 37–39, 62). Indeed, the models we consider here are based on similar logic.

    View all citing articles on Scopus
    View full text