Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(28): e2202370119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35749382

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections initiate in the bronchi of the upper respiratory tract and are able to disseminate to the lower respiratory tract, where infections can cause an acute respiratory distress syndrome with a high degree of mortality in elderly patients. We used reconstituted primary bronchial epithelia from adult and child donors to follow the SARS-CoV-2 infection dynamics. We show that, in epithelia from adult donors, infections initiate in multiciliated cells and spread within 24 to 48 h throughout the whole epithelia. Syncytia formed of ciliated and basal cells appeared at the apical side of the epithelia within 3 to 4 d and were released into the apical lumen, where they contributed to the transmittable virus dose. A small number of reconstituted epithelia were intrinsically more resistant to virus infection, limiting virus spread to different degrees. This phenotype was more frequent in epithelia derived from children versus adults and correlated with an accelerated release of type III interferon. Treatment of permissive adult epithelia with exogenous type III interferon restricted infection, while type III interferon gene knockout promoted infection. Furthermore, a transcript analysis revealed that the inflammatory response was specifically attenuated in children. Taken together, our findings suggest that apical syncytia formation is an underappreciated source of virus propagation for tissue or environmental dissemination, whereas a robust type III interferon response such as commonly seen in young donors restricted SARS-CoV-2 infection. Thus, the combination of interferon restriction and attenuated inflammatory response in children might explain the epidemiological observation of age-related susceptibility to COVID-19.


Subject(s)
Bronchi , COVID-19 , Giant Cells , Interferons , Respiratory Mucosa , SARS-CoV-2 , Aged , Bronchi/immunology , Bronchi/virology , COVID-19/immunology , COVID-19/virology , Child , Disease Susceptibility , Giant Cells/immunology , Giant Cells/virology , Humans , Interferons/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , SARS-CoV-2/immunology , Interferon Lambda
2.
Biochimie ; 195: 71-76, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34780840

ABSTRACT

As ZIKV continues to spread, many "unknowns" remain and research is needed to advance the understanding of this important pathogen. Viral RNA dependent-RNA polymerases (RdRp) are validated targets for inhibitors of the replication of several viruses. Several studies have set up in vitro enzymatic assays of the RdRp of the Zika virus for testing of candidate inhibitors. While most of these studies use short synthetic polymers, we have shown in a previous work that the Zika polymerase domain is capable of a de novo synthesis of the viral genome using the natural viral RNA as template. Here we have studied the role of the sequences at the 3'end of the minus-strand RNA in the initiation of the RNA synthesis by the Zika isolated RdRp. Our results strongly suggest that the region containing the 105 first nucleotides from the 3' end of the minus-strand RNA is important for initiation of the positive RNA synthesis. This indicates that this region displays all the primary and secondary structures to be efficiently recognized by the recombinant RdRp in vitro. Moreover, we show that the 46 nucleotides are sufficient to initiate RNA synthesis. In addition, the ZIKV polymerase domain poorly replicated the RNA of other RNA viruses and appeared highly selective for its own RNA.


Subject(s)
RNA-Dependent RNA Polymerase , Zika Virus Infection , Zika Virus , Humans , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Virus Replication , Zika Virus/enzymology , Zika Virus/genetics , Zika Virus/physiology
3.
Microorganisms ; 9(11)2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34835480

ABSTRACT

BACKGROUND: HIV infects around one hundred thousand patients in the Republic of the Congo. Approximately 25% of them receive an antiretroviral treatment; current first-line regimens include two NRTIs and one NNRTI, reverse transcriptase inhibitors. Recently, protease inhibitors (PIs) were also introduced as second-line therapy upon clinical signs of treatment failure. Due to the limited number of molecular characterizations and amount of drug resistance data available in the Republic of the Congo, this study aims to evaluate the prevalence of circulating resistance mutations within the pol region. METHODS: HIV-positive, ART-experienced patients have been enrolled in four semi-urban localities in the Republic of the Congo. Plasma samples were collected, and viral RNA was extracted. The viral load for each patient was evaluated by RT-qPCR, following the general diagnostic procedures of the University Hospital of Bordeaux. Finally, drug resistance genotyping and phylogenetic analysis were conducted following Sanger sequencing of the pol region. RESULTS: A high diversity of HIV-1 strains was observed with many recombinant forms. Drug resistance mutations in RT and PR genes were determined and correlated to HAART. Because integrase inhibitors are rarely included in treatments in the Republic of the Congo, the prevalence of integrase drug resistance mutations before treatment was also determined. Interestingly, very few mutations were observed. CONCLUSIONS: We confirmed a high diversity of HIV-1 in the Republic of the Congo. Most patients presented an accumulation of mutations conferring resistance against NRTIs, NNRTIs and PIs. Nonetheless, the absence of integrase mutations associated with drug resistance suggests that the introduction of integrase inhibitors into therapy will be highly beneficial to patients in the Republic of the Congo.

4.
Nucleic Acids Res ; 49(19): 11241-11256, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34634812

ABSTRACT

The stable insertion of the retroviral genome into the host chromosomes requires the association between integration complexes and cellular chromatin via the interaction between retroviral integrase and the nucleosomal target DNA. This final association may involve the chromatin-binding properties of both the retroviral integrase and its cellular cofactor LEDGF/p75. To investigate this and better understand the LEDGF/p75-mediated chromatin tethering of HIV-1 integrase, we used a combination of biochemical and chromosome-binding assays. Our study revealed that retroviral integrase has an intrinsic ability to bind and recognize specific chromatin regions in metaphase even in the absence of its cofactor. Furthermore, this integrase chromatin-binding property was modulated by the interaction with its cofactor LEDGF/p75, which redirected the enzyme to alternative chromosome regions. We also better determined the chromatin features recognized by each partner alone or within the functional intasome, as well as the chronology of efficient LEDGF/p75-mediated targeting of HIV-1 integrase to chromatin. Our data support a new chromatin-binding function of integrase acting in concert with LEDGF/p75 for the optimal association with the nucleosomal substrate. This work also provides additional information about the behavior of retroviral integration complexes in metaphase chromatin and the mechanism of action of LEDGF/p75 in this specific context.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chromatin/metabolism , HIV Integrase/genetics , Histones/genetics , Host-Pathogen Interactions/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , Chromatin/chemistry , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HIV Integrase/metabolism , Histones/metabolism , Humans , K562 Cells , Primary Cell Culture , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Transcription Factors/metabolism
5.
Molecules ; 26(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34500856

ABSTRACT

Multiple viral targets are now available in the clinic to fight HIV infection. Even if this targeted therapy is highly effective at suppressing viral replication, caregivers are facing growing therapeutic failures in patients due to resistance, with or without treatment-adherence glitches. Accordingly, it is important to better understand how HIV and other retroviruses replicate in order to propose alternative antiviral strategies. Recent studies have shown that multiple cellular factors are implicated during the integration step and, more specifically, that integrase can be regulated through post-translational modifications. We have shown that integrase is phosphorylated by GCN2, a cellular protein kinase of the integrated stress response, leading to a restriction of HIV replication. In addition, we found that this mechanism is conserved among other retroviruses. Accordingly, we developed an in vitro interaction assay, based on the AlphaLISA technology, to monitor the integrase-GCN2 interaction. From an initial library of 133 FDA-approved molecules, we identified nine compounds that either inhibited or stimulated the interaction between GCN2 and HIV integrase. In vitro characterization of these nine hits validated this pilot screen and demonstrated that the GCN2-integrase interaction could be a viable solution for targeting integrase out of its active site.


Subject(s)
HIV Infections/therapy , HIV Integrase/metabolism , Protein Serine-Threonine Kinases/metabolism , Small Molecule Libraries/chemistry , Virus Replication/drug effects , Catalytic Domain , Drug Evaluation, Preclinical , HIV , HIV Integrase/genetics , High-Throughput Screening Assays , Humans , Models, Molecular , Protein Binding , Protein Serine-Threonine Kinases/genetics , Retroviridae , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Virus Replication/genetics
6.
J Med Chem ; 64(12): 8579-8598, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34106711

ABSTRACT

Novel anti-HIV agents are still needed to overcome resistance issues, in particular inhibitors acting against novel viral targets. The ribonuclease H (RNase H) function of the reverse transcriptase (RT) represents a validated and promising target, and no inhibitor has reached the clinical pipeline yet. Here, we present rationally designed non-diketo acid selective RNase H inhibitors (RHIs) based on the quinolinone scaffold starting from former dual integrase (IN)/RNase H quinolinonyl diketo acids. Several derivatives were synthesized and tested against RNase H and viral replication and found active at micromolar concentrations. Docking studies within the RNase H catalytic site, coupled with site-directed mutagenesis, and Mg2+ titration experiments demonstrated that our compounds coordinate the Mg2+ cofactor and interact with amino acids of the RNase H domain that are highly conserved among naïve and treatment-experienced patients. In general, the new inhibitors influenced also the polymerase activity of RT but were selective against RNase H vs the IN enzyme.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/enzymology , Quinolones/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/metabolism , HeLa Cells , Humans , Magnesium/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , Protein Binding , Quinolones/chemical synthesis , Quinolones/metabolism , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/metabolism , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Virus Replication/drug effects
7.
ACS Med Chem Lett ; 11(5): 798-805, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435387

ABSTRACT

Due to the biological liability of diketo acid (DKA) chain, we transferred this element of our previously reported anti-HIV-1 pyrrolyl derivatives to a non-DKA scaffold, obtaining a series of pyrrolyl-pyrazole carboxylic acids as new RNase H inhibitors. Among the newly synthesized derivatives, oxyphenylpyrrolyl-pyrazoles demonstrated inhibitory activities within the low micromolar/submicromolar range with compound 11b being the most potent. Interestingly, all tested compounds showed up to 2 orders of magnitude of selectivity for RNase H vs integrase. Docking studies within the RNase H catalytic site, coupled with site-directed mutagenesis, showed the key structural features that could confer the ability to establish specific interactions within RNase H. Furthermore, they proved the ability of our compounds to interact with amino acids highly conserved among HIV-1 subspecies isolated among patients carrying drug-resistant variants. In the end, the newly discovered pyrazole carboxylic acid derivatives feature promising serum stability with respect to their corresponding DKAs.

8.
Molecules ; 24(20)2019 Oct 12.
Article in English | MEDLINE | ID: mdl-31614773

ABSTRACT

Currently, an increasing number of drugs are becoming available to clinics for the treatment of HIV infection. Even if this targeted therapy is highly effective at suppressing viral replication, caregivers are facing growing therapeutic failures in patients, due to resistance with or without treatment adherence concerns. Accordingly, it is important to continue to discover small molecules that have a novel mechanism of inhibition. In this work, HIV integrase inhibitors were selected by high-throughput screening. Chemical structure comparisons enabled the identification of stilbene disulfonic acids as a potential new chemotype. Biochemical characterization of the lead compound stilbenavir (NSC34931) and a few derivatives was performed. Stilbene disulfonic acid derivatives exhibit low to sub-micromolar antiviral activity, and they inhibit integrase through DNA-binding inhibition. They probably bind to the C-terminal domain of integrase, in the cavity normally occupied by the noncleaved strand of the viral DNA substrate. Because of this original mode of action compared to active site strand transfer inhibitors, they do not exhibit cross-resistance to the three main resistance pathways to integrase inhibitors (G140S-Q148H, N155H, and Y143R). Further structure-activity optimization should enable the development of more active and less toxic derivatives with potential clinical relevance.


Subject(s)
HIV Infections/drug therapy , HIV Integrase Inhibitors/chemistry , HIV Integrase/genetics , HIV/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Catalytic Domain/drug effects , Drug Resistance, Viral , HIV/enzymology , HIV/pathogenicity , HIV Infections/enzymology , HIV Infections/virology , HIV Integrase Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Mutation , Virus Replication/drug effects
9.
Molecules ; 23(8)2018 Jul 26.
Article in English | MEDLINE | ID: mdl-30049955

ABSTRACT

HIV-1 integrase (IN) inhibitors represent a new class of highly effective anti-AIDS therapeutics. Current FDA-approved IN strand transfer inhibitors (INSTIs) share a common mechanism of action that involves chelation of catalytic divalent metal ions. However, the emergence of IN mutants having reduced sensitivity to these inhibitors underlies efforts to derive agents that antagonize IN function by alternate mechanisms. Integrase along with the 96-residue multifunctional accessory protein, viral protein R (Vpr), are both components of the HIV-1 pre-integration complex (PIC). Coordinated interactions within the PIC are important for viral replication. Herein, we report a 7-mer peptide based on the shortened Vpr (69⁻75) sequence containing a biotin group and a photo-reactive benzoylphenylalanyl residue, and which exhibits low micromolar IN inhibitory potency. Photo-crosslinking experiments have indicated that the peptide directly binds IN. The peptide does not interfere with IN-DNA interactions or induce higher-order, aberrant IN multimerization, suggesting a mode of action for the peptide that is distinct from clinically used INSTIs and developmental allosteric IN inhibitors. This compact Vpr-derived peptide may serve as a valuable pharmacological tool to identify a potential new pharmacologic site.


Subject(s)
Gene Products, vpr/chemistry , Gene Products, vpr/metabolism , HIV Infections/virology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/physiology , Peptides/pharmacology , Amino Acid Sequence , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization
10.
J Med Chem ; 60(17): 7315-7332, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28737946

ABSTRACT

Integrase mutations can reduce the effectiveness of the first-generation FDA-approved integrase strand transfer inhibitors (INSTIs), raltegravir (RAL) and elvitegravir (EVG). The second-generation agent, dolutegravir (DTG), has enjoyed considerable clinical success; however, resistance-causing mutations that diminish the efficacy of DTG have appeared. Our current findings support and extend the substrate envelope concept that broadly effective INSTIs can be designed by filling the envelope defined by the DNA substrates. Previously, we explored 1-hydroxy-2-oxo-1,2-dihydro-1,8-naphthyridine-3-carboxamides as an INSTI scaffold, making a limited set of derivatives, and concluded that broadly effective INSTIs can be developed using this scaffold. Herein, we report an extended investigation of 6-substituents as well the first examples of 7-substituted analogues of this scaffold. While 7-substituents are not well-tolerated, we have identified novel substituents at the 6-position that are highly effective, with the best compound (6p) retaining better efficacy against a broad panel of known INSTI resistant mutants than any analogues we have previously described.


Subject(s)
HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Naphthyridines/chemistry , Naphthyridines/pharmacology , Cell Line , Crystallography, X-Ray , Drug Resistance, Viral , HIV Infections/drug therapy , HIV Infections/virology , HIV Integrase/chemistry , HIV Integrase/genetics , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Models, Molecular , Mutation , Virus Replication/drug effects
11.
Sci Rep ; 7(1): 2697, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28577343

ABSTRACT

Mosquito- and tick-borne pathogens including Chikungunya, Dengue, Japanese encephalitis, West Nile, Yellow fever and Zika virus, represent a new economic and public health challenge. In the absence of effective vaccines and specific therapies, only supportive regimens are administrated for most of these infections. Thus, the development of a targeted therapy is mandatory to stop the rapid progression of these pathogens and preoccupant associated burdens such as Guillain-Barre syndrome, microcephaly. For this, it is essential to develop biochemical tools to help study and target key viral enzymes involved in replication such as helicase complexes, methyl-transferases and RNA-dependent RNA polymerases. Here, we show that a highly purified ZIKV polymerase domain is active in vitro. Importantly, we show that this isolated domain is capable of de novo synthesis of the viral genome and efficient elongation without terminal nucleotide transferase activity. Altogether, this isolated polymerase domain will be a precious tool to screen and optimize specific nucleoside and non-nucleoside inhibitors to fight against Zika infections.


Subject(s)
RNA, Viral/genetics , RNA-Dependent RNA Polymerase/metabolism , Transcription, Genetic , Zika Virus Infection/virology , Zika Virus/physiology , Catalysis , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Protein Interaction Domains and Motifs , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , Virus Replication
12.
Nucleic Acids Res ; 44(14): 6896-906, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27369381

ABSTRACT

Integrase strand transfer inhibitors (INSTIs) are highly effective against HIV infections. Co-crystal structures of the prototype foamy virus intasome have shown that all three FDA-approved drugs, raltegravir (RAL), elvitegravir and dolutegravir (DTG), act as interfacial inhibitors during the strand transfer (ST) integration step. However, these structures give only a partial sense for the limited inhibition of the 3'-processing reaction by INSTIs and how INSTIs can be modified to overcome drug resistance, notably against the G140S-Q148H double mutation. Based on biochemical experiments with modified oligonucleotides, we demonstrate that both the viral DNA +1 and -1 bases, which flank the 3'-processing site, play a critical role for 3'-processing efficiency and inhibition by RAL and DTG. In addition, the G140S-Q148H (SH) mutant integrase, which has a reduced 3'-processing activity, becomes more active and more resistant to inhibition of 3'-processing by RAL and DTG in the absence of the -1 and +1 bases. Molecular modeling of HIV-1 integrase, together with biochemical data, indicate that the conserved residue Q146 in the flexible loop of HIV-1 integrase is critical for productive viral DNA binding through specific contacts with the virus DNA ends in the 3'-processing and ST reactions. The potency of integrase inhibitors against 3'-processing and their ability to overcome resistance is discussed.


Subject(s)
Catalytic Domain , DNA, Viral/metabolism , Drug Resistance, Viral/drug effects , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , Biocatalysis/drug effects , Guanine/metabolism , HIV Integrase/chemistry , HIV Integrase Inhibitors/chemistry , Ions , Magnesium/pharmacology , Models, Molecular , Mutation/genetics , Substrate Specificity/drug effects
13.
ACS Chem Biol ; 11(4): 1074-81, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26808478

ABSTRACT

HIV integrase (IN) strand transfer inhibitors (INSTIs) are among the newest anti-AIDS drugs; however, mutant forms of IN can confer resistance. We developed noncytotoxic naphthyridine-containing INSTIs that retain low nanomolar IC50 values against HIV-1 variants harboring all of the major INSTI-resistant mutations. We found by analyzing crystal structures of inhibitors bound to the IN from the prototype foamy virus (PFV) that the most successful inhibitors show striking mimicry of the bound viral DNA prior to 3'-processing and the bound host DNA prior to strand transfer. Using this concept of "bi-substrate mimicry," we developed a new broadly effective inhibitor that not only mimics aspects of both the bound target and viral DNA but also more completely fills the space they would normally occupy. Maximizing shape complementarity and recapitulating structural components encompassing both of the IN DNA substrates could serve as a guiding principle for the development of new INSTIs.


Subject(s)
Drug Resistance, Viral , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , Crystallography, X-Ray , HIV Integrase Inhibitors/chemistry
14.
Curr Top Med Chem ; 16(4): 435-40, 2016.
Article in English | MEDLINE | ID: mdl-26268341

ABSTRACT

Integrase (IN) is an essential viral enzyme required for HIV-1 replication, which has been targeted by anti-AIDS therapeutics. Integrase strand transfer inhibitors (INSTIs) represent a new class of antiretroviral agents developed for the treatment of HIV-1 infections. Important structural features that are shared by many INSTIs include a coplanar arrangement of three heteroatoms that chelate two catalytic Mg(2+) ions in the IN active site and a linked halophenyl ring that binds in the hydrophobic pocket formed by the complex of IN with viral DNA. We recently reported bicyclic 6,7-dihydroxyoxoisoindolin-1-one-based IN inhibitors. In the current study, we modified these inhibitors in three ways. First, we increased the spacer length between the metalchelating triad and the halophenyl group. Second, we replaced the indoline [5,6] bicycle with a fused dihydroxyisoquinolinones [6,6] ring system. Finally, we prepared bis-6,7-dihydroxyisoindolin-1-one-4-sulfonamides as dimeric HIV-1 IN inhibitors. These new analogues showed low micromolar inhibitory potency in in vitro HIV-1 integrase assays.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , Isoindoles/pharmacology , HIV Integrase Inhibitors/chemistry , HIV-1/drug effects , HIV-1/enzymology , Humans , Isoindoles/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
15.
Eur J Med Chem ; 104: 127-38, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26451771

ABSTRACT

The synthesis and antiviral evaluation of a series of dihydropyrimidinone and thiopyrimidine derivatives bearing aryl α,γ-diketobutanoic acid moiety are described using the Biginelli multicomponent reaction as key step. The most active among 20 synthesized novel compounds were 4c, 4d and 5b, which possess nanomolar HIV-1 integrase (IN) stand transfer (ST) inhibition activities. In order to understand their mode of interactions within the IN active site, we docked all the compounds into the previously reported X-ray crystal structure of IN. We observed that compounds 4c, 4d and 5b occupied an area close to the two catalytic Mg(2+) ions surrounded by their chelating triad (E221, D128 and D185), DC16, Y212 and the ß-diketo acid moiety of 4c, 4d and 5b chelating Mg(2+). As those compounds lack anti-HIV activities in cell, their prodrugs were synthetized. The prodrug 4c' exhibited an anti-HIV activity of 0.19 µM in primary human lymphocytes with some cytotoxicity. All together, these results indicate that the new analogs potentially interact within the catalytic site with highly conserved residues important for IN catalytic activity.


Subject(s)
Anti-HIV Agents/pharmacology , Butyric Acid/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Pyrimidines/pharmacology , Animals , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Butyric Acid/chemical synthesis , Butyric Acid/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Vero Cells
16.
Biochimie ; 118: 173-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363100

ABSTRACT

During clinical trials, a number of fully characterized molecules are dropped along the way because they do not provide enough benefit for the patient. Some of them show limited side effects and might be of great use for other applications. AS1411 is a nucleolin-targeting aptamer that underwent phase II clinical trials as anticancer agent. Here, we show that AS1411 exhibits extremely potent antiviral activity and is therefore an attractive new lead as anti-HIV agent.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Oligodeoxyribonucleotides/pharmacology , Antineoplastic Agents/pharmacology , Aptamers, Nucleotide , Cell Line , Cell Proliferation/drug effects , Humans
17.
J Med Chem ; 58(11): 4610-23, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25961960

ABSTRACT

Bifunctional quinolinonyl DKA derivatives were first described as nonselective inhibitors of 3'-processing (3'-P) and strand transfer (ST) functions of HIV-1 integrase (IN), while 7-aminosubstituted quinolinonyl derivatives were proven IN strand transfer inhibitors (INSTIs) that also displayed activity against ribonuclease H (RNase H). In this study, we describe the design, synthesis, and biological evaluation of new quinolinonyl diketo acid (DKA) derivatives characterized by variously substituted alkylating groups on the nitrogen atom of the quinolinone ring. Removal of the second DKA branch of bifunctional DKAs, and the amino group in position 7 of quinolinone ring combined with a fine-tuning of the substituents on the benzyl group in position 1 of the quinolinone, increased selectivity for IN ST activity. In vitro, the most potent compound was 11j (IC50 = 10 nM), while the most active compounds against HIV infected cells were ester derivatives 10j and 10l. In general, the activity against RNase H was negligible, with only a few compounds active at concentrations higher than 10 µM. The binding mode of the most potent IN inhibitor 11j within the IN catalytic core domain (CCD) is described as well as its binding mode within the RNase H catalytic site to rationalize its selectivity.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/chemistry , Keto Acids/pharmacology , Quinolones/pharmacology , RNA-Directed DNA Polymerase/chemistry , Ribonuclease H/antagonists & inhibitors , Catalytic Domain , HIV Infections/drug therapy , HIV Infections/virology , HIV Integrase Inhibitors/chemistry , HIV-1/drug effects , HeLa Cells , Humans , Keto Acids/chemistry , Models, Molecular , Molecular Structure , Quinolones/chemistry , Structure-Activity Relationship , Virus Replication/drug effects
18.
J Med Chem ; 58(4): 1915-28, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25629256

ABSTRACT

The development of HIV-1 dual inhibitors is a highly innovative approach aimed at reducing drug toxic side effects as well as therapeutic costs. HIV-1 integrase (IN) and reverse transcriptase-associated ribonuclease H (RNase H) are both selective targets for HIV-1 chemotherapy, and the identification of dual IN/RNase H inhibitors is an attractive strategy for new drug development. We newly synthesized pyrrolyl derivatives that exhibited good potency against IN and a moderate inhibition of the RNase H function of RT, confirming the possibility of developing dual HIV-1 IN/RNase H inhibitors and obtaining new information for the further development of more effective dual HIV-1 inhibitors.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , HIV/drug effects , Pyrroles/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H/antagonists & inhibitors , Dose-Response Relationship, Drug , HIV/enzymology , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , Microbial Sensitivity Tests , Molecular Structure , Protein Structure, Tertiary/drug effects , Pyrroles/chemical synthesis , Pyrroles/chemistry , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Ribonuclease H/metabolism , Structure-Activity Relationship , Virus Replication/drug effects
19.
Nucleic Acids Res ; 42(20): 12352-66, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25332402

ABSTRACT

G-rich nucleic acids can form non-canonical G-quadruplex structures (G4s) in which four guanines fold in a planar arrangement through Hoogsteen hydrogen bonds. Although many biochemical and structural studies have focused on DNA sequences containing successive, adjacent guanines that spontaneously fold into G4s, evidence for their in vivo relevance has recently begun to accumulate. Complete sequencing of the human genome highlighted the presence of ∼300,000 sequences that can potentially form G4s. Likewise, the presence of putative G4-sequences has been reported in various viruses genomes [e.g., Human immunodeficiency virus (HIV-1), Epstein-Barr virus (EBV), papillomavirus (HPV)]. Many studies have focused on telomeric G4s and how their dynamics are regulated to enable telomere synthesis. Moreover, a role for G4s has been proposed in cellular and viral replication, recombination and gene expression control. In parallel, DNA aptamers that form G4s have been described as inhibitors and diagnostic tools to detect viruses [e.g., hepatitis A virus (HAV), EBV, cauliflower mosaic virus (CaMV), severe acute respiratory syndrome virus (SARS), simian virus 40 (SV40)]. Here, special emphasis will be given to the possible role of these structures in a virus life cycle as well as the use of G4-forming oligonucleotides as potential antiviral agents and innovative tools.


Subject(s)
Antiviral Agents/chemistry , DNA, Viral/chemistry , G-Quadruplexes , RNA, Viral/chemistry , Genome, Human , Humans
20.
J Med Chem ; 57(12): 5190-202, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24901667

ABSTRACT

There are currently three HIV-1 integrase (IN) strand transfer inhibitors (INSTIs) approved by the FDA for the treatment of AIDS. However, the emergence of drug-resistant mutants emphasizes the need to develop additional agents that have improved efficacies against the existent resistant mutants. As reported herein, we modified our recently disclosed 1-hydroxy-2-oxo-1,2-dihydro-1,8-naphthyridine-3-carboxamides IN inhibitors to develop compounds that have improved efficacies against recombinant IN in biochemical assays. These new compounds show single-digit nanomolar antiviral potencies against HIV vectors that carry wild-type (WT) IN in a single round replication assay and have improved potency against vectors harboring the major forms of drug resistant IN mutants. These compounds also have low toxicity for cultured cells, which in several cases, results in selectivity indices (CC50/EC50) of greater than 10000. The compounds have the potential, with additional structural modifications, to yield clinical agents that are effective against the known strains of resistant viruses.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/genetics , HIV-1/drug effects , Naphthyridines/pharmacology , Pyrrolidinones/pharmacology , Cell Line, Tumor , Drug Resistance, Viral , HEK293 Cells , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , HIV-1/enzymology , HIV-1/genetics , Humans , Mutation , Naphthyridines/chemical synthesis , Naphthyridines/chemistry , Raltegravir Potassium , Recombinant Proteins/chemistry , Stereoisomerism , Structure-Activity Relationship , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...