Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Functional and Structural Characterization of 2-Amino-4-phenylthiazole Inhibitors of the HIV-1 Nucleocapsid Protein with Antiviral Activity
My Activity
  • Open Access
Letters

Functional and Structural Characterization of 2-Amino-4-phenylthiazole Inhibitors of the HIV-1 Nucleocapsid Protein with Antiviral Activity
Click to copy article linkArticle link copied!

View Author Information
Department of Biotechnology, Chemistry and Pharmacy, Università degli Studi di Siena, via Aldo Moro 2, I-53019 Siena, Italy
Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, UMR 7213 CNRS, 74 route du Rhin, F-67401 Illkirch, France
§ Laboratoire de Cristallographie et RMN Biologiques, CNRS, Paris Sorbonne Cité, 4 avenue de l’Observatoire, F-75006 Paris, France
Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, D-60596 Frankfurt, Germany
Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, BioLife Science Bldg., Suite 333, 1900 N. 12th Street, Philadelphia, Pennsylvania 19122, United States
Open PDFSupporting Information (1)

ACS Chemical Biology

Cite this: ACS Chem. Biol. 2014, 9, 9, 1950–1955
Click to copy citationCitation copied!
https://doi.org/10.1021/cb500316h
Published July 2, 2014

Copyright © 2014 American Chemical Society. This publication is licensed under these Terms of Use.

Abstract

Click to copy section linkSection link copied!

The nucleocapsid protein (NC) is a highly conserved protein in diverse HIV-1 subtypes that plays a central role in virus replication, mainly by interacting with conserved nucleic acid sequences. NC is considered a highly profitable drug target to inhibit multiple steps in the HIV-1 life cycle with just one compound, a unique property not shown by any of the other antiretroviral classes. However, most of NC inhibitors developed so far act through an unspecific and potentially toxic mechanism (zinc ejection) and are mainly being investigated as topical microbicides. In an effort to provide specific NC inhibitors that compete for the binding of nucleic acids to NC, here we combined molecular modeling, organic synthesis, biophysical studies, NMR spectroscopy, and antiviral assays to design, synthesize, and characterize an efficient NC inhibitor endowed with antiviral activity in vitro, a desirable property for the development of efficient antiretroviral lead compounds.

Copyright © 2014 American Chemical Society

During the past 25 years, the arsenal of drugs to combat HIV-1 infection has increased continuously, even though most anti-HIV-1 drugs experienced clinical failure due to antiretroviral drug resistance resulting from mutations in HIV-1 protein targets as a consequence of the error-prone nature of HIV-1 reverse transcriptase (RT) and its lack of a proofreading function. The most common and realistic strategy adopted so far to overcome this issue is the design of more potent and selective inhibitors of entry/fusion, RT, Integrase (IN), and Protease (PR), but these compounds often lack full activity against drug-resistant virus strains selected by the same drug classes. (1) Therefore, a medical need in this field is still felt, and additional conserved drug targets need to be explored to achieve virus suppression in drug-experienced patients or patients infected with drug-resistant viruses. In this context, the HIV-1 nucleocapsid protein (NC), which is highly conserved throughout different HIV-1 subtypes and exerts essential functions in virus replication, is a promising anti-HIV-1 target. (2, 3) NC is a 55-amino-acid small basic protein with two zinc fingers that assists RT during reverse transcription by chaperoning the annealing of the cellular primer tRNA to the primer binding site (PBS) and the two obligatory DNA strand transfers necessary for the synthesis of a complete functional double-stranded vDNA with two long terminal repeats. (4) NC is also thought to protect the nascent vDNA against nucleases (5) and to assist IN for the integration of the viral DNA into the host genome. (6) As a domain of the Gag structural polyprotein precursor, NC selects the genomic RNA and promotes its dimerization and packaging during virus assembly. (7-9) Finally, NC binding to RNA is critical for Gag processing by PR and for the subsequent formation of the condensed ribonucleoprotein architecture within the virion. (10) NC can hardly mutate, so that NC mutants are generally nonfunctional and result in noninfectious viruses. (11, 12) Therefore, specific inhibition of NC is thought to generate a sustained antiretroviral activity.

Inhibition of NC has been achieved by different strategies leading to antiretroviral effects in vitro and in vivo, (13) although generally through unspecific mechanisms and thus causing significant cytotoxicity. Indeed, there is a critical deficiency of small molecule NC inhibitors (NCIs) endowed with a clear and safe mechanism of action as well as with antiviral activity in vitro, which may be developed as effective and safe antiretroviral agents.

Targeting protein/nucleic acids interaction by small molecules is a highly promising strategy in anti-HIV therapy, even if generally considered as a tough challenge due to several difficulties, most often related to the intrinsic structural disorder and highly basic nature of nucleic acids-binding proteins. In continuation of our interest in discovering specific NCIs competing with nucleic acids for the binding to NC and exhibiting antiviral activity, we established a multidisciplinary strategy based on three pioneer works in the field. Indeed, Shvadchak et al. have developed a high-throughput screening assay based on targeting the NC chaperone activities, which led to the identification of five molecular fragments that efficiently inhibit NC but do not show antiviral activity. (14) Recently, Goudreau and co-workers have provided the first solution structure of NC in complex with an NCI that mimics the guanosine found in many NC/RNA complexes. (15) Moreover, as NC is a highly flexible protein, it was essential to use molecular dynamics (MD) simulations to study the conformational behavior of NC in complex with nucleic acids and to develop a new strategy based on virtual screening to design small molecule NCIs showing antiretroviral activity. (16, 17)

On the basis of a combination of molecular simulations, biophysical studies, NMR analysis, and antiviral assays, here we elucidate the NC inhibitory activity and structural requirements for NC binding of AN3, a new NCI targeting efficiently NC nucleic acid chaperone activity and endowed with antiviral activity in cells. Notably, AN3 was designed starting from NCI fragments identified by Shvadchak, (14) whose complexes with NC have been already investigated by molecular docking simulations and GRID-based analysis. (16) Starting from docking complexes, here we performed preliminary MD simulations of 25 ns each, showing that fragment A10 had the highest conformational stability within the hydrophobic pocket of NC located at the top of the folded zinc fingers (Supporting Information, Figure S1), which play a key role in NC chaperoning activities. (18-20) For this reason, A10 was selected for further studies aimed at corroborating the validity of the 2-amino-4-phenylthiazole scaffold for NC inhibition as well as to develop NCI with antiviral activity in cells.

In agreement with our previous observations, (16) MD analysis showed that the aminothiazole moiety nicely fits the hydrophobic pocket of NC and may behave as a guanine-mimetic. The catechol phenyl ring is stacked with the Trp37 side chain, whereas hydroxyl groups are projected toward the solvent but do not bind to basic residues that have been shown to play a key role in binding to nucleic acids. (21) Moreover, catechol species are particularly sensitive to oxidation, giving quinone or semiquinone radicals, which may interfere with the functional characterization of the mechanism of action through binding to proteins or nucleic acids. Indeed, a number of catechols of polyhydroxylated compounds have been previously shown to inhibit NC functions by an unclear mechanism, but without showing antiviral activity in vitro. (22) Therefore, our strategy focused on the structure-guided replacement of the catechol moiety in A10 with functional groups that may eventually improve membrane permeability. To this end, compounds sharing the common 2-amino-4-phenylthiazole substructure and bearing substitutions on the phenyl ring were downloaded from the ZINC database (about 950 molecules) and submitted to the virtual screening protocol already described. (16) In parallel, these molecules were also docked toward the NMR structure of NC in complex with an NCI. (15) Compounds AN1, AN2, and AN3 (Figure 1A and Supporting Information, Table S1, for basic properties) were prioritized by virtual screening and further studied by means of 0.3 μs of MD for checking their conformational stability (Supporting Information). Then, these molecules were purchased and tested in vitro.

We first monitored the effect of AN compounds on the NC(11-55)-induced destabilization of the secondary structure of cTAR DNA, which is exquisitely dependent on the hydrophobic plateau at the top of the properly folded zinc finger motifs, (19) by using the double-labeled Rh6G-5′-cTAR-3′-Dabcyl derivative. In the absence of NC, cTAR is mainly in a nonfluorescent closed form, where the Rh6G and Dabcyl labels at the 5′ and 3′ termini of the cTAR stem are close together. Addition of a 10-fold excess of NC(11-55) led to melting of the bottom of the cTAR stem, partially restoring the Rh6G fluorescence. (23) Therefore, NCIs designed to interfere with NC/nucleic acid interactions are expected to reverse the NC-promoted increase in Rh6G fluorescence. Addition of 100 μM concentration of AN1, AN2, and AN3 to the NC(11-55)/cTAR complex was found to induce 7%, 10%, and 28% decrease of the Rh6G emission, respectively (Figure 1B). A control experiment performed in the absence of NC excluded any direct effect of the compounds on Rh6G emission (data not shown). Thus, all three compounds are able to partially inhibit the NC(11-55)-induced destabilization of cTAR, AN3 being the most efficient NCI with an IC50 of 140(±90) μM (Figure 1B, insert). The inhibition constant Ki was then calculated according to the Cheng and Prussof equation:(1)with [cTAR] = 100 nM and the dissociation constant (Kd(NC-cTAR) = 5.9 × 10–8 M) of NC(11-55) for cTAR. (19) A Ki value of 50(±30) μM was obtained for AN3.

Figure 1

Figure 1. AN compounds and their effects on the NC(11-55)-induced cTAR destabilization. (A) Chemical structure of A10 and the three compounds selected by virtual screening. (B) Emission spectra of cTAR (0.1 μM) were recorded in the absence (black) and in the presence of 1 μM NC(11-55) before (red) and after addition of 100 μM AN1 (blue), AN2 (magenta), or AN3 (green). Excitation wavelength was at 520 nm. The concentration of DMSO was less than 2% by volume. Insert: Inhibition of NC(11-55)-induced cTAR melting by AN3. The percentage of NC(11-55) activity is plotted as a function of the AN3 concentration. An IC50 value of 140 ± 90 μM was calculated for AN3 from the fit of the experimental data (squares) to the following equation: % NC-induced cTAR destabilization = 1/(1 + (IC50/[AN3])p), where p = 1.8(±1) is the Hill coefficient.

Since AN3 showed the most efficient inhibition of NC chaperone activity, it was selected for further characterization and synthesized in larger quantities as outlined in Scheme 1. Compound 3 was obtained by nucleophilic substitution of commercially available methyl 4-(bromomethyl)benzoate 1 with 2′-hydroxyacetophenone 2 in a very good yield. Bromination at the α position of the ketone with copper(II) bromide afforded the monohalogenated compound 4 in moderate yield (60%); this latter was cyclized with thiourea in N,N-dimethylformamide to afford the 2-aminothiazole 5 quantitatively. (24) Finally the methyl ester was hydrolyzed with lithium hydroxide to afford AN3.

Scheme 1

Scheme 1. Synthesis of AN3a

Scheme aReaction and conditions: (i) K2CO3, DMF, rt, overnight; (ii) CuBr2, CHCl3/AcOEt (1:1), reflux, overnight; (iii) thiourea, DMF, rt, 1 h; (iv) LiOH·H2O, THF/MeOH/H2O (3:1:1), rt 5 h.

To confirm the direct binding of AN3 to NC as well as to rule out possible zinc ejection, we analyzed by mass spectrometry the changes in the mass of NC(11-55) after addition of AN3 to NC(11-55) at 1:1, 2:1, 5:1, and 10:1 molar ratio. While no peak indicates that AN3 could eject zinc, a peak corresponding to the sum of the masses of NC(11-55) and AN3 indicates the formation of a 1:1 complex with NC(11-55) (Supporting Information, Figure S2). Notably, the adduct between the reference fragment A10 and NC(11-55) was not detected by MS analysis. (14)

In a next step, the binding constant of AN3 to NC(11-55) was measured by monitoring the changes in the fluorescence anisotropy of AN3, which is intrinsically fluorescent, during its titration by NC(11-55) (Supporting Information, Figure S3). Assuming a 1:1 stoichiometry, we found an association constant KNC-AN3 of (1.0 ± 0.2) × 104 M–1, which is well consistent with the Ki value determined in Figure 1B. We further characterized the binding properties of AN3 to NC(11-55) by isothermal titration calorimetry (ITC). Since marginal heat exchange accompanied the binding of AN3 to NC(11-55) even at high concentrations of both partners, indicating a mainly entropy-driven interaction, we performed a competition titration in which AN3 competed with cTAR for binding to NC(11-55). By monitoring the heat of reaction associated with the binding of NC to cTAR (Supporting Information, Figure S4), we obtained an association constant KNC-AN3 = (1.4 ± 0.3) × 104 M–1, in good agreement with the above value.

Moreover, the ITC experiment confirmed that AN3 is a competitive inhibitor of NC/nucleic acids interaction. Taken together, our experiments consistently indicated that AN3 binds to NC(11-55) with a Kd value of ∼100 μM.

To check whether AN3 may directly interact with the Trp37 residue, which has been shown to be crucial for NC activity, (4, 19, 21, 25) we monitored the intrinsic fluorescence emission of the Trp37 residue in NC(11-55) upon the addition of 100 μM AN3, a concentration close to its Kd value. (26, 27)AN3 binding was found to decrease by 22% the fluorescence emission of Trp37, in line with a direct binding of AN3 to the Trp37 residue (Supporting Information, Figure S5). This direct interaction was also observed by molecular modeling, as the phenyl-aminothiazole moiety was found to be persistently over the Trp37 side chain along MD trajectories (Figure 2A). This nearly stacked conformation may well explain the decrease of Trp37 fluorescence emission, since Trp37 fluorescence was shown to be highly sensitive to stacking with oligonucleotide bases. (26, 28)

Figure 2

Figure 2. Binding of AN3 to NC. (A) Representative MD structure: AN3 is shown as cyan sticks, NC as cartoon and transparent surface. Residues highlighted by NMR and biophysical studies are shown as sticks and colored orange. (B) AN3 protons involved in the interaction with NC by STD NMR experiment are shown on the STD spectrum. AN3 concentration was 1 mM, whereas NC concentration was 30 μM. The STD experiment was recorded with selective saturation of the methyl group resonances of NC at 1 ppm, and with selective saturation in an empty spectral region (off-resonance). The subtraction of the spectra was performed internally via phase cycling after every scan to minimize the subtraction artifacts. The resulting subtracted spectrum contains only the signals of AN3 interacting with NC. The signals of NC were suppressed with the use of a transverse relaxation filter applied before the acquisition.

NMR saturation-transfer difference (STD) (29) and chemical shift mapping experiments further confirmed the interaction of AN3 with NC. STD experiment (Figure 2B) clearly indicates that H5, H7, and the equivalent H16 and H18 protons of AN3 are involved in the interaction with NC. Moreover, NMR chemical shift mapping performed on N15-labeled NC suggests that residues Ala25, Asn17, and Met46 (as well as Gly4, Gln45, Cys18, Lys38, albeit to a lesser extent) of NC are involved in the binding with AN3 (Supporting Information, Figure S6). These findings nicely match with the binding mode of AN3 addressed by MD simulations (Figure 2A), thus facilitating further structure-based computer-aided ligand optimization studies. In particular, chemical optimization of AN3 should concentrate on improving intermolecular interactions with the NC to enhance its inhibitory potency, as well as optimizing the druggability of the small molecule. Concentrations of DMSO (50%) necessary for solubilizing AN3 during the NMR chemical shift mapping experiment prevented us from observing the interaction of AN3 with Trp37. Most probably, DMSO competed with AN3 for hydrophobic interactions with NC.

Antiretroviral activity against HIV-1 and cytotoxicity of AN3 were preliminarily evaluated as previously described. (17, 30) Briefly, HeLa P4.R5MAGI cells (expressing CD4, CCR5, and β-gal under the control of HIV-1 LTR) were infected by HIV-1Lai (MOI = 0.3) in the presence of serial dilutions of AN3 in medium for 24 h. After extensive washing, fresh medium with the respective AN3 concentrations was added. Viral supernatants were collected 24 h later, and viral titers were quantified on TZM-bl cells expressing firefly luciferase from an HIV-1 LTR promoter. Luciferase activity in cell lysates was measured by luminometry 44 h after infection. Compared to the DMSO control, which was set to 100% infection, AN3 clearly showed concentration-dependent antiviral activity with EC50 = 95 μM (Figure 3). This value is highly comparable to the Kd value calculated by ITC or fluorescence anisotropy, as well as to the IC50 value determined for the inhibition of NC(11-55)-induced cTAR destabilization, strongly suggesting that NC may be the primary target of the antiviral activity of AN3. Notably, complete inhibition was always achieved at 250 μM, a concentration where toxic effects were still low (25%) (Supporting Information, Figure S7).

Figure 3

Figure 3. AN3 showed a concentration-dependent antiviral activity in vitro. Infectious viruses produced by P4.R5MAGI cells in the presence of AN3 were quantified by luminometry on Tzm-bl cells. The mean of triplicates is shown with standard deviations. Data were normalized with respect to DMSO (100%). One of two representative experiments is shown.

In conclusion, starting from the previously identified NCI fragment A10, here we designed AN3 as an efficient inhibitor of the HIV-1 NC chaperone functions. Although the NC inhibitory activities of A10 and AN3 are comparable, AN3 showed antiviral activity in infected cells with a mechanism of action clearly indicating NC as the primary target. Structural requirements for AN3 binding to NC were elucidated by NMR, biophysical studies, and MD simulations, showing that AN3 interacts within the hydrophobic pocket of NC and competes with nucleic acids for this binding site. In agreement with MD simulations, a direct interaction of AN3 to the key residue Trp37 was observed, whereas NMR chemical shift mapping experiments further highlighted the involvement of Met46, Ala25, and Asn17, thus clearly identifying the hydrophobic pocket of NC as the binding site of AN3. Our multidisciplinary strategy demonstrates that the challenging goal of targeting the NC with specific small molecules is truly feasible, thus paving the way for the structure-based design of effective NCI. Overall, AN3 fulfills all expectations for an effective NCI, thus becoming a valuable starting point for further medicinal chemistry-oriented optimization.

Supporting Information

Click to copy section linkSection link copied!

Molecular dynamics, materials, mass spectrometry, general biophysics directions, isothermal titration calorimetry, general NMR directions, chemistry, and details of the antiretroviral and cytotoxicity assays. This material is available free of charge via the Internet at http://pubs.acs.org.

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Authors
    • Yves Mely - Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, UMR 7213 CNRS, 74 route du Rhin, F-67401 Illkirch, France Email: [email protected]
    • Maurizio Botta - Department of Biotechnology, Chemistry and Pharmacy, Università degli Studi di Siena, via Aldo Moro 2, I-53019 Siena, ItalySbarro Institute for Cancer Research and Molecular Medicine, Temple University, BioLife Science Bldg., Suite 333, 1900 N. 12th Street, Philadelphia, Pennsylvania 19122, United States Email: [email protected]
  • Authors
    • Mattia Mori - Department of Biotechnology, Chemistry and Pharmacy, Università degli Studi di Siena, via Aldo Moro 2, I-53019 Siena, Italy
    • Alessandro Nucci - Department of Biotechnology, Chemistry and Pharmacy, Università degli Studi di Siena, via Aldo Moro 2, I-53019 Siena, Italy
    • Maria Chiara Dasso Lang - Department of Biotechnology, Chemistry and Pharmacy, Università degli Studi di Siena, via Aldo Moro 2, I-53019 Siena, Italy
    • Nicolas Humbert - Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, UMR 7213 CNRS, 74 route du Rhin, F-67401 Illkirch, France
    • Christian Boudier - Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, UMR 7213 CNRS, 74 route du Rhin, F-67401 Illkirch, France
    • Francois Debaene - Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, UMR 7213 CNRS, 74 route du Rhin, F-67401 Illkirch, France
    • Sarah Sanglier-Cianferani - Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, UMR 7213 CNRS, 74 route du Rhin, F-67401 Illkirch, France
    • Marjorie Catala - Laboratoire de Cristallographie et RMN Biologiques, CNRS, Paris Sorbonne Cité, 4 avenue de l’Observatoire, F-75006 Paris, France
    • Patricia Schult-Dietrich - Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, D-60596 Frankfurt, Germany
    • Ursula Dietrich - Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, D-60596 Frankfurt, Germany
    • Carine Tisné - Laboratoire de Cristallographie et RMN Biologiques, CNRS, Paris Sorbonne Cité, 4 avenue de l’Observatoire, F-75006 Paris, France
  • Notes
    The authors declare no competing financial interest.

Acknowledgment

Click to copy section linkSection link copied!

This work was supported by the European Project THINPAD “Targeting the HIV-1 Nucleocapsid Protein to fight Antiretroviral Drug Resistance” (FP7–Grant Agreement 601969).

References

Click to copy section linkSection link copied!

This article references 30 other publications.

  1. 1
    Tang, M. W. and Shafer, R. W. (2012) HIV-1 antiretroviral resistance: scientific principles and clinical applications Drugs 72, e1 25
  2. 2
    Darlix, J. L., Godet, J., Ivanyi-Nagy, R., Fosse, P., Mauffret, O., and Mely, Y. (2011) Flexible nature and specific functions of the HIV-1 nucleocapsid protein J. Mol. Biol. 410, 565 581
  3. 3
    Thomas, J. A. and Gorelick, R. J. (2008) Nucleocapsid protein function in early infection processes Virus Res. 134, 39 63
  4. 4
    Godet, J. and Mely, Y. (2010) Biophysical studies of the nucleic acid chaperone properties of the HIV-1 nucleocapsid protein RNA Biol. 7, 687 699
  5. 5
    Krishnamoorthy, G., Roques, B., Darlix, J. L., and Mely, Y. (2003) DNA condensation by the nucleocapsid protein of HIV-1: a mechanism ensuring DNA protection Nucleic Acids Res. 31, 5425 5432
  6. 6
    Poljak, L., Batson, S. M., Ficheux, D., Roques, B. P., Darlix, J. L., and Kas, E. (2003) Analysis of NCp7-dependent activation of HIV-1 cDNA integration and its conservation among retroviral nucleocapsid proteins J. Mol. Biol. 329, 411 421
  7. 7
    Zhang, Y., Qian, H., Love, Z., and Barklis, E. (1998) Analysis of the assembly function of the human immunodeficiency virus type 1 gag protein nucleocapsid domain J. Virol. 72, 1782 1789
  8. 8
    Muriaux, D., Mirro, J., Harvin, D., and Rein, A. (2001) RNA is a structural element in retrovirus particles Proc. Natl. Acad. Sci. U.S.A. 98, 5246 5251
  9. 9
    Clever, J., Sassetti, C., and Parslow, T. G. (1995) RNA secondary structure and binding sites for gag gene products in the 5′ packaging signal of human immunodeficiency virus type 1 J. Virol. 69, 2101 2109
  10. 10
    Mirambeau, G., Lyonnais, S., Coulaud, D., Hameau, L., Lafosse, S., Jeusset, J., Borde, I., Reboud-Ravaux, M., Restle, T., Gorelick, R. J., and Le Cam, E. (2007) HIV-1 protease and reverse transcriptase control the architecture of their nucleocapsid partner PLoS One 2, e669
  11. 11
    Demene, H., Dong, C. Z., Ottmann, M., Rouyez, M. C., Jullian, N., Morellet, N., Mely, Y., Darlix, J. L., Fournie-Zaluski, M. C., and Saragosti, S. 1994, 1H NMR structure and biological studies of the His23—>Cys mutant nucleocapsid protein of HIV-1 indicate that the conformation of the first zinc finger is critical for virus infectivity Biochemistry 33, 11707 11716
  12. 12
    Gorelick, R. J., Gagliardi, T. D., Bosche, W. J., Wiltrout, T. A., Coren, L. V., Chabot, D. J., Lifson, J. D., Henderson, L. E., and Arthur, L. O. (1999) Strict conservation of the retroviral nucleocapsid protein zinc finger is strongly influenced by its role in viral infection processes: characterization of HIV-1 particles containing mutant nucleocapsid zinc-coordinating sequences Virology 256, 92 104
  13. 13
    Mori, M., Manetti, F., and Botta, M. (2011) Targeting protein-protein and protein-nucleic acid interactions for anti-HIV therapy Curr. Pharm. Des. 17, 3713 3728
  14. 14
    Shvadchak, V., Sanglier, S., Rocle, S., Villa, P., Haiech, J., Hibert, M., Van Dorsselaer, A., Mely, Y., and de Rocquigny, H. (2009) Identification by high throughput screening of small compounds inhibiting the nucleic acid destabilization activity of the HIV-1 nucleocapsid protein Biochimie 91, 916 923
  15. 15
    Goudreau, N., Hucke, O., Faucher, A. M., Grand-Maitre, C., Lepage, O., Bonneau, P. R., Mason, S. W., and Titolo, S. (2013) Discovery and structural characterization of a new inhibitor series of HIV-1 nucleocapsid function: NMR solution structure determination of a ternary complex involving a 2:1 inhibitor/NC stoichiometry J. Mol. Biol. 425, 1982 1998
  16. 16
    Mori, M., Manetti, F., and Botta, M. (2011) Predicting the binding mode of known NCp7 inhibitors to facilitate the design of novel modulators J. Chem. Inf. Model. 51, 446 454
  17. 17
    Mori, M., Schult-Dietrich, P., Szafarowicz, B., Humbert, N., Debaene, F., Sanglier-Cianferani, S., Dietrich, U., Mely, Y., and Botta, M. (2012) Use of virtual screening for discovering antiretroviral compounds interacting with the HIV-1 nucleocapsid protein Virus Res. 169, 377 387
  18. 18
    Avilov, S. V., Piemont, E., Shvadchak, V., de Rocquigny, H., and Mely, Y. (2008) Probing dynamics of HIV-1 nucleocapsid protein/target hexanucleotide complexes by 2-aminopurine Nucleic Acids Res. 36, 885 896
  19. 19
    Beltz, H., Clauss, C., Piemont, E., Ficheux, D., Gorelick, R. J., Roques, B., Gabus, C., Darlix, J. L., de Rocquigny, H., and Mely, Y. (2005) Structural determinants of HIV-1 nucleocapsid protein for cTAR DNA binding and destabilization, and correlation with inhibition of self-primed DNA synthesis J. Mol. Biol. 348, 1113 1126
  20. 20
    Godet, J., Ramalanjaona, N., Sharma, K. K., Richert, L., de Rocquigny, H., Darlix, J. L., Duportail, G., and Mely, Y. (2011) Specific implications of the HIV-1 nucleocapsid zinc fingers in the annealing of the primer binding site complementary sequences during the obligatory plus strand transfer Nucleic Acids Res. 39, 6633 6645
  21. 21
    Mori, M., Dietrich, U., Manetti, F., and Botta, M. (2010) Molecular dynamics and DFT study on HIV-1 nucleocapsid protein-7 in complex with viral genome J. Chem. Inf. Model. 50, 638 650
  22. 22
    Cruceanu, M., Stephen, A. G., Beuning, P. J., Gorelick, R. J., Fisher, R. J., and Williams, M. C. (2006) Single DNA molecule stretching measures the activity of chemicals that target the HIV-1 nucleocapsid protein Anal. Biochem. 358, 159 170
  23. 23
    Bernacchi, S. and Mely, Y. (2001) Exciton interaction in molecular beacons: a sensitive sensor for short range modifications of the nucleic acid structure Nucleic Acids Res. 29, E62 62
  24. 24
    Zav’yalov, S. I., Kravchenko, N. E., Ezhova, G. I., Kulikova, L. B., Zavozin, A. G., and Dorofeeva, O. V. (2007) Synthesis of 2-aminothiazole derivatives Pharm. Chem. J. 41, 105 108
  25. 25
    De Guzman, R. N., Wu, Z. R., Stalling, C. C., Pappalardo, L., Borer, P. N., and Summers, M. F. (1998) Structure of the HIV-1 nucleocapsid protein bound to the SL3 psi-RNA recognition element Science 279, 384 388
  26. 26
    Mely, Y., Piemont, E., Sorinas-Jimeno, M., de Rocquigny, H., Jullian, N., Morellet, N., Roques, B. P., and Gerard, D. (1993) Structural and dynamic characterization of the aromatic amino acids of the human immunodeficiency virus type I nucleocapsid protein zinc fingers and their involvement in heterologous tRNA(Phe) binding: a steady-state and time-resolved fluorescence study Biophys. J. 65, 1513 1522
  27. 27
    Mely, Y., De Rocquigny, H., Morellet, N., Roques, B. P., and Gerad, D. (1996) Zinc binding to the HIV-1 nucleocapsid protein: a thermodynamic investigation by fluorescence spectroscopy Biochemistry 35, 5175 5182
  28. 28
    Vuilleumier, C., Bombarda, E., Morellet, N., Gerard, D., Roques, B. P., and Mely, Y. (1999) Nucleic acid sequence discrimination by the HIV-1 nucleocapsid protein NCp7: a fluorescence study Biochemistry 38, 16816 16825
  29. 29
    Mayer, M. and Meyer, B. (2001) Group epitope mapping by saturation transfer difference NMR to identify segments of a ligand in direct contact with a protein receptor J. Am. Chem. Soc. 123, 6108 6117
  30. 30
    Dietz, J., Koch, J., Kaur, A., Raja, C., Stein, S., Grez, M., Pustowka, A., Mensch, S., Ferner, J., Möller, L., Bannert, N., Tampe, R., Divita, G., Mely, Y., Schwalbe, H., and Dietrich, U. (2008) Inhibition of HIV-1 by a peptide ligand of the genomic RNA packaging signal Psi ChemMedChem. 3, 749 755

Cited By

Click to copy section linkSection link copied!

This article is cited by 24 publications.

  1. Savina Malancona, Mattia Mori, Paola Fezzardi, Marisabella Santoriello, Andreina Basta, Martina Nibbio, Lesia Kovalenko, Roberto Speziale, Maria Rosaria Battista, Antonella Cellucci, Nadia Gennari, Edith Monteagudo, Annalise Di Marco, Alessia Giannini, Rajhans Sharma, Manuel Pires, Eleonore Real, Maurizio Zazzi, Maria Chiara Dasso Lang, Davide De Forni, Francesco Saladini, Yves Mely, Vincenzo Summa, Steven Harper, Maurizio Botta. 5,6-Dihydroxypyrimidine Scaffold to Target HIV-1 Nucleocapsid Protein. ACS Medicinal Chemistry Letters 2020, 11 (5) , 766-772. https://doi.org/10.1021/acsmedchemlett.9b00608
  2. Nicolas Humbert, Lesia Kovalenko, Francesco Saladini, Alessia Giannini, Manuel Pires, Thomas Botzanowski, Sergiy Cherenok, Christian Boudier, Kamal K. Sharma, Eleonore Real, Olga A. Zaporozhets, Sarah Cianférani, Carole Seguin-Devaux, Federica Poggialini, Maurizio Botta, Maurizio Zazzi, Vitaly I. Kalchenko, Mattia Mori, Yves Mély. (Thia)calixarenephosphonic Acids as Potent Inhibitors of the Nucleic Acid Chaperone Activity of the HIV-1 Nucleocapsid Protein with a New Binding Mode and Multitarget Antiviral Activity. ACS Infectious Diseases 2020, 6 (4) , 687-702. https://doi.org/10.1021/acsinfecdis.9b00290
  3. Mattia Mori, Fabrizio Manetti, Bruno Botta, Andrea Tafi. In Memory of Maurizio Botta: His Contribution to the Development of Computer-Aided Drug Design. Journal of Chemical Information and Modeling 2019, 59 (12) , 4961-4967. https://doi.org/10.1021/acs.jcim.9b01043
  4. Mattia Mori, Maria Chiara Dasso Lang, Francesco Saladini, Nastasja Palombi, Lesia Kovalenko, Davide De Forni, Barbara Poddesu, Laura Friggeri, Alessia Giannini, Savina Malancona, Vincenzo Summa, Maurizio Zazzi, Yves Mely, Maurizio Botta. Synthesis and Evaluation of Bifunctional Aminothiazoles as Antiretrovirals Targeting the HIV-1 Nucleocapsid Protein. ACS Medicinal Chemistry Letters 2019, 10 (4) , 463-468. https://doi.org/10.1021/acsmedchemlett.8b00506
  5. Mattia Mori, Lesia Kovalenko, Savina Malancona, Francesco Saladini, Davide De Forni, Manuel Pires, Nicolas Humbert, Eleonore Real, Thomas Botzanowski, Sarah Cianférani, Alessia Giannini, Maria Chiara Dasso Lang, Giulia Cugia, Barbara Poddesu, Franco Lori, Maurizio Zazzi, Steven Harper, Vincenzo Summa, Yves Mely, and Maurizio Botta . Structure-Based Identification of HIV-1 Nucleocapsid Protein Inhibitors Active against Wild-Type and Drug-Resistant HIV-1 Strains. ACS Chemical Biology 2018, 13 (1) , 253-266. https://doi.org/10.1021/acschembio.7b00907
  6. Peng Zhan, Christophe Pannecouque, Erik De Clercq, and Xinyong Liu . Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends. Journal of Medicinal Chemistry 2016, 59 (7) , 2849-2878. https://doi.org/10.1021/acs.jmedchem.5b00497
  7. Virginia Pontecorvi, Mattia Mori, Francesca Picarazzi, Susi Zara, Simone Carradori, Amelia Cataldi, Andrea Angeli, Emanuela Berrino, Paola Chimenti, Alessia Ciogli, Daniela Secci, Paolo Guglielmi, Claudiu T. Supuran. Novel Insights on Human Carbonic Anhydrase Inhibitors Based on Coumalic Acid: Design, Synthesis, Molecular Modeling Investigation, and Biological Studies. International Journal of Molecular Sciences 2022, 23 (14) , 7950. https://doi.org/10.3390/ijms23147950
  8. Atilio Reyes Romero, Serjey Lunev, Grzegorz M. Popowicz, Vito Calderone, Matteo Gentili, Michael Sattler, Jacek Plewka, Michał Taube, Maciej Kozak, Tad A. Holak, Alexander S. S. Dömling, Matthew R. Groves. A fragment-based approach identifies an allosteric pocket that impacts malate dehydrogenase activity. Communications Biology 2021, 4 (1) https://doi.org/10.1038/s42003-021-02442-1
  9. Mattia Mori, Stefano Ciaco, Yves Mély, Anastasia Karioti. Inhibitory Effect of Lithospermic Acid on the HIV-1 Nucleocapsid Protein. Molecules 2020, 25 (22) , 5434. https://doi.org/10.3390/molecules25225434
  10. Julia Menzel, Daniel Kownatzki-Danger, Sergiy Tokar, Alice Ballone, Kirsten Unthan-Fechner, Markus Kilisch, Christof Lenz, Henning Urlaub, Mattia Mori, Christian Ottmann, Michael J. Shattock, Stephan E. Lehnart, Blanche Schwappach. 14-3-3 binding creates a memory of kinase action by stabilizing the modified state of phospholamban. Science Signaling 2020, 13 (647) https://doi.org/10.1126/scisignal.aaz1436
  11. Alexej Dick, Simon Cocklin. Recent Advances in HIV-1 Gag Inhibitor Design and Development. Molecules 2020, 25 (7) , 1687. https://doi.org/10.3390/molecules25071687
  12. Khalid M. Al‐Anazi, Ahmed H. Mahmoud, Mohammad AbulFarah, Ahmed A. Allam, Moustafa M. G. Fouda, Hatem E. Gaffer. 2‐Amino‐5‐arylazothiazole‐Based Derivatives: In Vitro Cytotoxicity, Antioxidant Properties, and Bleomycin‐Dependent DNA Damage. ChemistrySelect 2019, 4 (19) , 5570-5576. https://doi.org/10.1002/slct.201901148
  13. J A Morales-Morales, A F Villamarin, E Florez-López, J J Rios-Acevedo. Study of the electrochemical oxidation of 2-Thiazolamine and 2-Oxazolamine on a platinum wire as working electrode. Journal of Physics: Conference Series 2018, 1119 , 012006. https://doi.org/10.1088/1742-6596/1119/1/012006
  14. J.A Morales-Morales, J. J. Rios-Acevedo, E. Florez-Lopez. A preliminary study of cyclic voltammetry in glassy carbon electrode. Redox Behaviour of 2-Amino-4-phenylthiazoles. Journal of Physics: Conference Series 2018, 1119 , 012016. https://doi.org/10.1088/1742-6596/1119/1/012016
  15. J A Morales-Morales, M Salazar-Osorio, E Florez López. Electrochemical reduction processes on glassy carbon electrode of 2-Amino-4-phenylthiazoles.. Journal of Physics: Conference Series 2018, 1119 , 012025. https://doi.org/10.1088/1742-6596/1119/1/012025
  16. Nunzio Iraci, Oriana Tabarrini, Claudio Santi, Luca Sancineto. NCp7: targeting a multitask protein for next-generation anti-HIV drug development part 2. Noncovalent inhibitors and nucleic acid binders. Drug Discovery Today 2018, 23 (3) , 687-695. https://doi.org/10.1016/j.drudis.2018.01.022
  17. Elia Gamba, Mattia Mori, Lesia Kovalenko, Alessia Giannini, Alice Sosic, Francesco Saladini, Dan Fabris, Yves Mély, Barbara Gatto, Maurizio Botta. Identification of novel 2-benzoxazolinone derivatives with specific inhibitory activity against the HIV-1 nucleocapsid protein. European Journal of Medicinal Chemistry 2018, 145 , 154-164. https://doi.org/10.1016/j.ejmech.2017.12.073
  18. Andrea Calcaterra, Valentina Iovine, Bruno Botta, Deborah Quaglio, Ilaria D’Acquarica, Alessia Ciogli, Antonia Iazzetti, Romina Alfonsi, Ludovica Lospinoso Severini, Paola Infante, Lucia Di Marcotullio, Mattia Mori, Francesca Ghirga. Chemical, computational and functional insights into the chemical stability of the Hedgehog pathway inhibitor GANT61. Journal of Enzyme Inhibition and Medicinal Chemistry 2018, 33 (1) , 349-358. https://doi.org/10.1080/14756366.2017.1419221
  19. Anand Balupuri, Dae-Yon Lee, Myeong Hwi Lee, Sangeun Chae, Eunmi Jung, Yunki Kim, Jeonghee Ryu, Nam Sook Kang. Design, synthesis, docking and biological evaluation of 4-phenyl-thiazole derivatives as autotaxin (ATX) inhibitors. Bioorganic & Medicinal Chemistry Letters 2017, 27 (17) , 4156-4164. https://doi.org/10.1016/j.bmcl.2017.07.022
  20. Jean-Luc Darlix, Hugues de Rocquigny, Yves Mély. The multiple roles of the nucleocapsid in retroviral RNA conversion into proviral DNA by reverse transcriptase. Biochemical Society Transactions 2016, 44 (5) , 1427-1440. https://doi.org/10.1042/BST20160101-T
  21. Christopher R. M. Asquith, Lidia S. Konstantinova, Tuomo Laitinen, Marina L. Meli, Antti Poso, Oleg A. Rakitin, Regina Hofmann‐Lehmann, Stephen T. Hilton. Evaluation of Substituted 1,2,3‐Dithiazoles as Inhibitors of the Feline Immunodeficiency Virus (FIV) Nucleocapsid Protein via a Proposed Zinc Ejection Mechanism. ChemMedChem 2016, 11 (19) , 2119-2126. https://doi.org/10.1002/cmdc.201600260
  22. Nithi Phukan, Jubaraj B. Baruah. Anion Guided Conformational Adjustments by Protonation Leading to Conformation Reversal. ChemistrySelect 2016, 1 (3) , 440-443. https://doi.org/10.1002/slct.201600020
  23. Honnappa Nagarajaiah, Abhaya Kumar Mishra, Jarugu Narasimha Moorthy. Mechanochemical solid-state synthesis of 2-aminothiazoles, quinoxalines and benzoylbenzofurans from ketones by one-pot sequential acid- and base-mediated reactions. Organic & Biomolecular Chemistry 2016, 14 (17) , 4129-4135. https://doi.org/10.1039/C6OB00351F
  24. Mattia Mori, Lesia Kovalenko, Sébastien Lyonnais, Danny Antaki, Bruce E. Torbett, Maurizio Botta, Gilles Mirambeau, Yves Mély. Nucleocapsid Protein: A Desirable Target for Future Therapies Against HIV-1. 2015, 53-92. https://doi.org/10.1007/82_2015_433
Open PDF

ACS Chemical Biology

Cite this: ACS Chem. Biol. 2014, 9, 9, 1950–1955
Click to copy citationCitation copied!
https://doi.org/10.1021/cb500316h
Published July 2, 2014

Copyright © 2014 American Chemical Society. This publication is licensed under these Terms of Use.

Article Views

1630

Altmetric

-

Citations

Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. AN compounds and their effects on the NC(11-55)-induced cTAR destabilization. (A) Chemical structure of A10 and the three compounds selected by virtual screening. (B) Emission spectra of cTAR (0.1 μM) were recorded in the absence (black) and in the presence of 1 μM NC(11-55) before (red) and after addition of 100 μM AN1 (blue), AN2 (magenta), or AN3 (green). Excitation wavelength was at 520 nm. The concentration of DMSO was less than 2% by volume. Insert: Inhibition of NC(11-55)-induced cTAR melting by AN3. The percentage of NC(11-55) activity is plotted as a function of the AN3 concentration. An IC50 value of 140 ± 90 μM was calculated for AN3 from the fit of the experimental data (squares) to the following equation: % NC-induced cTAR destabilization = 1/(1 + (IC50/[AN3])p), where p = 1.8(±1) is the Hill coefficient.

    Scheme 1

    Scheme 1. Synthesis of AN3a

    Scheme aReaction and conditions: (i) K2CO3, DMF, rt, overnight; (ii) CuBr2, CHCl3/AcOEt (1:1), reflux, overnight; (iii) thiourea, DMF, rt, 1 h; (iv) LiOH·H2O, THF/MeOH/H2O (3:1:1), rt 5 h.

    Figure 2

    Figure 2. Binding of AN3 to NC. (A) Representative MD structure: AN3 is shown as cyan sticks, NC as cartoon and transparent surface. Residues highlighted by NMR and biophysical studies are shown as sticks and colored orange. (B) AN3 protons involved in the interaction with NC by STD NMR experiment are shown on the STD spectrum. AN3 concentration was 1 mM, whereas NC concentration was 30 μM. The STD experiment was recorded with selective saturation of the methyl group resonances of NC at 1 ppm, and with selective saturation in an empty spectral region (off-resonance). The subtraction of the spectra was performed internally via phase cycling after every scan to minimize the subtraction artifacts. The resulting subtracted spectrum contains only the signals of AN3 interacting with NC. The signals of NC were suppressed with the use of a transverse relaxation filter applied before the acquisition.

    Figure 3

    Figure 3. AN3 showed a concentration-dependent antiviral activity in vitro. Infectious viruses produced by P4.R5MAGI cells in the presence of AN3 were quantified by luminometry on Tzm-bl cells. The mean of triplicates is shown with standard deviations. Data were normalized with respect to DMSO (100%). One of two representative experiments is shown.

  • References


    This article references 30 other publications.

    1. 1
      Tang, M. W. and Shafer, R. W. (2012) HIV-1 antiretroviral resistance: scientific principles and clinical applications Drugs 72, e1 25
    2. 2
      Darlix, J. L., Godet, J., Ivanyi-Nagy, R., Fosse, P., Mauffret, O., and Mely, Y. (2011) Flexible nature and specific functions of the HIV-1 nucleocapsid protein J. Mol. Biol. 410, 565 581
    3. 3
      Thomas, J. A. and Gorelick, R. J. (2008) Nucleocapsid protein function in early infection processes Virus Res. 134, 39 63
    4. 4
      Godet, J. and Mely, Y. (2010) Biophysical studies of the nucleic acid chaperone properties of the HIV-1 nucleocapsid protein RNA Biol. 7, 687 699
    5. 5
      Krishnamoorthy, G., Roques, B., Darlix, J. L., and Mely, Y. (2003) DNA condensation by the nucleocapsid protein of HIV-1: a mechanism ensuring DNA protection Nucleic Acids Res. 31, 5425 5432
    6. 6
      Poljak, L., Batson, S. M., Ficheux, D., Roques, B. P., Darlix, J. L., and Kas, E. (2003) Analysis of NCp7-dependent activation of HIV-1 cDNA integration and its conservation among retroviral nucleocapsid proteins J. Mol. Biol. 329, 411 421
    7. 7
      Zhang, Y., Qian, H., Love, Z., and Barklis, E. (1998) Analysis of the assembly function of the human immunodeficiency virus type 1 gag protein nucleocapsid domain J. Virol. 72, 1782 1789
    8. 8
      Muriaux, D., Mirro, J., Harvin, D., and Rein, A. (2001) RNA is a structural element in retrovirus particles Proc. Natl. Acad. Sci. U.S.A. 98, 5246 5251
    9. 9
      Clever, J., Sassetti, C., and Parslow, T. G. (1995) RNA secondary structure and binding sites for gag gene products in the 5′ packaging signal of human immunodeficiency virus type 1 J. Virol. 69, 2101 2109
    10. 10
      Mirambeau, G., Lyonnais, S., Coulaud, D., Hameau, L., Lafosse, S., Jeusset, J., Borde, I., Reboud-Ravaux, M., Restle, T., Gorelick, R. J., and Le Cam, E. (2007) HIV-1 protease and reverse transcriptase control the architecture of their nucleocapsid partner PLoS One 2, e669
    11. 11
      Demene, H., Dong, C. Z., Ottmann, M., Rouyez, M. C., Jullian, N., Morellet, N., Mely, Y., Darlix, J. L., Fournie-Zaluski, M. C., and Saragosti, S. 1994, 1H NMR structure and biological studies of the His23—>Cys mutant nucleocapsid protein of HIV-1 indicate that the conformation of the first zinc finger is critical for virus infectivity Biochemistry 33, 11707 11716
    12. 12
      Gorelick, R. J., Gagliardi, T. D., Bosche, W. J., Wiltrout, T. A., Coren, L. V., Chabot, D. J., Lifson, J. D., Henderson, L. E., and Arthur, L. O. (1999) Strict conservation of the retroviral nucleocapsid protein zinc finger is strongly influenced by its role in viral infection processes: characterization of HIV-1 particles containing mutant nucleocapsid zinc-coordinating sequences Virology 256, 92 104
    13. 13
      Mori, M., Manetti, F., and Botta, M. (2011) Targeting protein-protein and protein-nucleic acid interactions for anti-HIV therapy Curr. Pharm. Des. 17, 3713 3728
    14. 14
      Shvadchak, V., Sanglier, S., Rocle, S., Villa, P., Haiech, J., Hibert, M., Van Dorsselaer, A., Mely, Y., and de Rocquigny, H. (2009) Identification by high throughput screening of small compounds inhibiting the nucleic acid destabilization activity of the HIV-1 nucleocapsid protein Biochimie 91, 916 923
    15. 15
      Goudreau, N., Hucke, O., Faucher, A. M., Grand-Maitre, C., Lepage, O., Bonneau, P. R., Mason, S. W., and Titolo, S. (2013) Discovery and structural characterization of a new inhibitor series of HIV-1 nucleocapsid function: NMR solution structure determination of a ternary complex involving a 2:1 inhibitor/NC stoichiometry J. Mol. Biol. 425, 1982 1998
    16. 16
      Mori, M., Manetti, F., and Botta, M. (2011) Predicting the binding mode of known NCp7 inhibitors to facilitate the design of novel modulators J. Chem. Inf. Model. 51, 446 454
    17. 17
      Mori, M., Schult-Dietrich, P., Szafarowicz, B., Humbert, N., Debaene, F., Sanglier-Cianferani, S., Dietrich, U., Mely, Y., and Botta, M. (2012) Use of virtual screening for discovering antiretroviral compounds interacting with the HIV-1 nucleocapsid protein Virus Res. 169, 377 387
    18. 18
      Avilov, S. V., Piemont, E., Shvadchak, V., de Rocquigny, H., and Mely, Y. (2008) Probing dynamics of HIV-1 nucleocapsid protein/target hexanucleotide complexes by 2-aminopurine Nucleic Acids Res. 36, 885 896
    19. 19
      Beltz, H., Clauss, C., Piemont, E., Ficheux, D., Gorelick, R. J., Roques, B., Gabus, C., Darlix, J. L., de Rocquigny, H., and Mely, Y. (2005) Structural determinants of HIV-1 nucleocapsid protein for cTAR DNA binding and destabilization, and correlation with inhibition of self-primed DNA synthesis J. Mol. Biol. 348, 1113 1126
    20. 20
      Godet, J., Ramalanjaona, N., Sharma, K. K., Richert, L., de Rocquigny, H., Darlix, J. L., Duportail, G., and Mely, Y. (2011) Specific implications of the HIV-1 nucleocapsid zinc fingers in the annealing of the primer binding site complementary sequences during the obligatory plus strand transfer Nucleic Acids Res. 39, 6633 6645
    21. 21
      Mori, M., Dietrich, U., Manetti, F., and Botta, M. (2010) Molecular dynamics and DFT study on HIV-1 nucleocapsid protein-7 in complex with viral genome J. Chem. Inf. Model. 50, 638 650
    22. 22
      Cruceanu, M., Stephen, A. G., Beuning, P. J., Gorelick, R. J., Fisher, R. J., and Williams, M. C. (2006) Single DNA molecule stretching measures the activity of chemicals that target the HIV-1 nucleocapsid protein Anal. Biochem. 358, 159 170
    23. 23
      Bernacchi, S. and Mely, Y. (2001) Exciton interaction in molecular beacons: a sensitive sensor for short range modifications of the nucleic acid structure Nucleic Acids Res. 29, E62 62
    24. 24
      Zav’yalov, S. I., Kravchenko, N. E., Ezhova, G. I., Kulikova, L. B., Zavozin, A. G., and Dorofeeva, O. V. (2007) Synthesis of 2-aminothiazole derivatives Pharm. Chem. J. 41, 105 108
    25. 25
      De Guzman, R. N., Wu, Z. R., Stalling, C. C., Pappalardo, L., Borer, P. N., and Summers, M. F. (1998) Structure of the HIV-1 nucleocapsid protein bound to the SL3 psi-RNA recognition element Science 279, 384 388
    26. 26
      Mely, Y., Piemont, E., Sorinas-Jimeno, M., de Rocquigny, H., Jullian, N., Morellet, N., Roques, B. P., and Gerard, D. (1993) Structural and dynamic characterization of the aromatic amino acids of the human immunodeficiency virus type I nucleocapsid protein zinc fingers and their involvement in heterologous tRNA(Phe) binding: a steady-state and time-resolved fluorescence study Biophys. J. 65, 1513 1522
    27. 27
      Mely, Y., De Rocquigny, H., Morellet, N., Roques, B. P., and Gerad, D. (1996) Zinc binding to the HIV-1 nucleocapsid protein: a thermodynamic investigation by fluorescence spectroscopy Biochemistry 35, 5175 5182
    28. 28
      Vuilleumier, C., Bombarda, E., Morellet, N., Gerard, D., Roques, B. P., and Mely, Y. (1999) Nucleic acid sequence discrimination by the HIV-1 nucleocapsid protein NCp7: a fluorescence study Biochemistry 38, 16816 16825
    29. 29
      Mayer, M. and Meyer, B. (2001) Group epitope mapping by saturation transfer difference NMR to identify segments of a ligand in direct contact with a protein receptor J. Am. Chem. Soc. 123, 6108 6117
    30. 30
      Dietz, J., Koch, J., Kaur, A., Raja, C., Stein, S., Grez, M., Pustowka, A., Mensch, S., Ferner, J., Möller, L., Bannert, N., Tampe, R., Divita, G., Mely, Y., Schwalbe, H., and Dietrich, U. (2008) Inhibition of HIV-1 by a peptide ligand of the genomic RNA packaging signal Psi ChemMedChem. 3, 749 755
  • Supporting Information

    Supporting Information


    Molecular dynamics, materials, mass spectrometry, general biophysics directions, isothermal titration calorimetry, general NMR directions, chemistry, and details of the antiretroviral and cytotoxicity assays. This material is available free of charge via the Internet at http://pubs.acs.org.


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.