Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(8): 102208, 2022 08.
Article in English | MEDLINE | ID: mdl-35772496

ABSTRACT

Antibiotic resistance via epigenetic methylation of ribosomal RNA is one of the most prevalent strategies adopted by multidrug resistant pathogens. The erythromycin-resistance methyltransferase (Erm) methylates rRNA at the conserved A2058 position and imparts resistance to macrolides such as erythromycin. However, the precise mechanism adopted by Erm methyltransferases for locating the target base within a complicated rRNA scaffold remains unclear. Here, we show that a conserved RNA architecture, including specific bulge sites, present more than 15 Å from the reaction center, is key to methylation at the pathogenic site. Using a set of RNA sequences site-specifically labeled by fluorescent nucleotide surrogates, we show that base flipping is a prerequisite for effective methylation and that distal bases assist in the recognition and flipping at the reaction center. The Erm-RNA complex model revealed that intrinsically flipped-out bases in the RNA serve as a putative anchor point for the Erm. Molecular dynamic simulation studies demonstrated the RNA undergoes a substantial change in conformation to facilitate an effective protein-rRNA handshake. This study highlights the importance of unique architectural features exploited by RNA to impart fidelity to RNA methyltransferases via enabling allosteric crosstalk. Moreover, the distal trigger sites identified here serve as attractive hotspots for the development of combination drug therapy aimed at reversing resistance.


Subject(s)
Methyltransferases , RNA, Ribosomal , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Drug Resistance, Microbial/genetics , Erythromycin/pharmacology , Methyltransferases/metabolism , RNA , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism
2.
Nucleic Acids Res ; 49(19): e111, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34450653

ABSTRACT

Interconversions between nucleic acid structures play an important role in transcriptional and translational regulation and also in repair and recombination. These interconversions are frequently promoted by nucleic acid chaperone proteins. To monitor their kinetics, Förster resonance energy transfer (FRET) is widely exploited using ensemble fluorescence intensity measurements in pre-steady-state stopped-flow experiments. Such experiments only provide a weighted average of the emission of all species in solution and consume large quantities of materials. Herein, we lift these limitations by combining time-resolved fluorescence (TRF) with droplet microfluidics (DmF). We validate the innovative TRF-DmF approach by investigating the well characterized annealing of the HIV-1 (+)/(-) Primer Binding Sequences (PBS) promoted by a HIV-1 nucleocapsid peptide. Upon rapid mixing of the FRET-labelled (-)PBS with its complementary (+)PBS sequence inside microdroplets, the TRF-DmF set-up enables resolving the time evolution of sub-populations of reacting species and reveals an early intermediate with a ∼50 ps donor fluorescence lifetime never identified so far. TRF-DmF also favorably compares with single molecule experiments, as it offers an accurate control of concentrations with no upper limit, no need to graft one partner on a surface and no photobleaching issues.


Subject(s)
DNA Primers/chemistry , HIV-1/chemistry , Molecular Chaperones/chemistry , Nucleocapsid Proteins/chemistry , Peptides/chemistry , Serum Albumin, Human/chemistry , Base Pairing , DNA Primers/metabolism , Fluoresceins/chemistry , Fluorescence , Fluorescence Resonance Energy Transfer , HIV-1/metabolism , Humans , Kinetics , Microfluidic Analytical Techniques , Molecular Chaperones/metabolism , Nucleic Acid Conformation , Nucleocapsid Proteins/metabolism , Peptides/metabolism , Serum Albumin, Human/metabolism , p-Dimethylaminoazobenzene/analogs & derivatives , p-Dimethylaminoazobenzene/chemistry
3.
J Am Chem Soc ; 142(40): 16999-17014, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32915558

ABSTRACT

Thienoguanosine (thG) is an isomorphic guanosine (G) surrogate that almost perfectly mimics G in nucleic acids. To exploit its full potential and lay the foundation for future applications, 20 DNA duplexes, where the bases facing and neighboring thG were systematically varied, were thoroughly studied using fluorescence spectroscopy, molecular dynamics simulations, and mixed quantum mechanical/molecular mechanics calculations, yielding a comprehensive understanding of its photophysics in DNA. In matched duplexes, thG's hypochromism was larger for flanking G/C residues but its fluorescence quantum yield (QY) and lifetime values were almost independent of the flanking bases. This was attributed to high duplex stability, which maintains a steady orientation and distance between nucleobases, so that a similar charge transfer (CT) mechanism governs the photophysics of thG independently of its flanking nucleobases. thG can therefore replace any G residue in matched duplexes, while always maintaining similar photophysical features. In contrast, the local destabilization induced by a mismatch or an abasic site restores a strong dependence of thG's QY and lifetime values on its environmental context, depending on the CT route efficiency and solvent exposure of thG. Due to this exquisite sensitivity, thG appears ideal for monitoring local structural changes and single nucleotide polymorphism. Moreover, thG's dominant fluorescence lifetime in DNA is unusually long (9-29 ns), facilitating its selective measurement in complex media using a lifetime-based or a time-gated detection scheme. Taken together, our data highlight thG as an outstanding emissive substitute for G with good QY, long fluorescence lifetimes, and exquisite sensitivity to local structural changes.


Subject(s)
DNA Probes/chemistry , DNA/chemistry , Fluorescent Dyes/chemistry , Guanosine/analogs & derivatives , Guanosine/chemistry , Kinetics , Molecular Dynamics Simulation , Nucleic Acid Conformation , Solvents/chemistry , Spectrometry, Fluorescence , Structure-Activity Relationship
4.
Nucleic Acids Res ; 48(16): 9218-9234, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32797159

ABSTRACT

The HIV-1 Gag protein playing a key role in HIV-1 viral assembly has recently been shown to interact through its nucleocapsid domain with the ribosomal protein L7 (RPL7) that acts as a cellular co-factor promoting Gag's nucleic acid (NA) chaperone activity. To further understand how the two proteins act together, we examined their mechanism individually and in concert to promote the annealing between dTAR, the DNA version of the viral transactivation element and its complementary cTAR sequence, taken as model HIV-1 sequences. Gag alone or complexed with RPL7 was found to act as a NA chaperone that destabilizes cTAR stem-loop and promotes its annealing with dTAR through the stem ends via a two-step pathway. In contrast, RPL7 alone acts as a NA annealer that through its NA aggregating properties promotes cTAR/dTAR annealing via two parallel pathways. Remarkably, in contrast to the isolated proteins, their complex promoted efficiently the annealing of cTAR with highly stable dTAR mutants. This was confirmed by the RPL7-promoted boost of the physiologically relevant Gag-chaperoned annealing of (+)PBS RNA to the highly stable tRNALys3 primer, favoring the notion that Gag recruits RPL7 to overcome major roadblocks in viral assembly.


Subject(s)
HIV Infections/genetics , HIV-1/genetics , Ribosomal Proteins/genetics , gag Gene Products, Human Immunodeficiency Virus/genetics , Amino Acid Sequence/genetics , HIV Infections/virology , HIV-1/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Molecular Chaperones/genetics , Nucleic Acid Conformation , Nucleic Acids/genetics , RNA, Viral/genetics , Virus Assembly/genetics
5.
ACS Med Chem Lett ; 11(5): 698-705, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435373

ABSTRACT

The HIV-1 nucleocapsid protein 7 (NC) is a potential target for effective antiretroviral therapy due to its central role in virus replication, mainly linked to nucleic acid (NA) chaperone activity, and low susceptibility to drug resistance. By screening a compounds library, we identified the aminopyrrolic compound CN14_17, a known carbohydrate binding agent, that inhibits the NC chaperone activity in the low micromolar range. Different from most of available NC inhibitors, CN14_17 fully prevents the NC-induced annealing of complementary NA sequences. Using fluorescence assays and isothermal titration calorimetry, we found that CN14_17 competes with NC for the binding to NAs, preferentially targeting single-stranded sequences. Molecular dynamics simulations confirmed that binding to cTAR occurs preferably within the guanosine-rich single stranded sequence. Finally, CN14_17 exhibited antiretroviral activity in the low micromolar range, although with a moderate therapeutic index. Overall, CN14_17 might be the progenitor of a new promising class of NC inhibitors.

6.
ACS Infect Dis ; 6(4): 687-702, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32045204

ABSTRACT

The nucleocapsid protein (NC) is a highly conserved protein that plays key roles in HIV-1 replication through its nucleic acid chaperone properties mediated by its two zinc fingers and basic residues. NC is a promising target for antiviral therapy, particularly to control viral strains resistant to currently available drugs. Since calixarenes with antiviral properties have been described, we explored the ability of calixarene hydroxymethylphosphonic or sulfonic acids to inhibit NC chaperone properties and exhibit antiviral activity. By using fluorescence-based assays, we selected four calixarenes inhibiting NC chaperone activity with submicromolar IC50 values. These compounds were further shown by mass spectrometry, isothermal titration calorimetry, and fluorescence anisotropy to bind NC with no zinc ejection and to compete with nucleic acids for the binding to NC. Molecular dynamic simulations further indicated that these compounds interact via their phosphonate or sulfonate groups with the basic surface of NC but not with the hydrophobic plateau at the top of the folded fingers. Cellular studies showed that the most soluble compound CIP201 inhibited the infectivity of wild-type and drug-resistant HIV-1 strains at low micromolar concentrations, primarily targeting the early steps of HIV-1 replication. Moreover, CIP201 was also found to inhibit the flipping and polymerization activity of reverse transcriptase. Calixarenes thus form a class of noncovalent NC inhibitors, endowed with a new binding mode and multitarget antiviral activity.


Subject(s)
Anti-HIV Agents/pharmacology , Calixarenes/pharmacology , HIV-1/chemistry , Molecular Chaperones/antagonists & inhibitors , Nucleocapsid Proteins/antagonists & inhibitors , Organophosphonates/pharmacology , Animals , Calixarenes/classification , HIV-1/drug effects , Inhibitory Concentration 50 , Mice , Mice, Transgenic , Molecular Dynamics Simulation , Protein Binding
7.
Chemistry ; 25(58): 13363-13375, 2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31322780

ABSTRACT

During DNA replication, ubiquitin-like, containing PHD and RING fingers domains 1 (UHRF1) plays key roles in the inheritance of methylation patterns to daughter strands by recognizing through its SET and RING-associated domain (SRA) the methylated CpGs and recruiting DNA methyltransferase 1 (DNMT1). Herein, our goal is to identify UHRF1 inhibitors targeting the 5'-methylcytosine (5mC) binding pocket of the SRA domain to prevent the recognition and flipping of 5mC and determine the molecular and cellular consequences of this inhibition. For this, we used a multidisciplinary strategy combining virtual screening and molecular modeling with biophysical assays in solution and cells. We identified an anthraquinone compound able to bind to the 5mC binding pocket and inhibit the base-flipping process in the low micromolar range. We also showed in cells that this hit impaired the UHRF1/DNMT1 interaction and decreased the overall methylation of DNA, highlighting the critical role of base flipping for DNMT1 recruitment and providing the first proof of concept of the druggability of the 5mC binding pocket. The selected anthraquinone appears thus as a key tool to investigate the role of UHRF1 in the inheritance of methylation patterns, as well as a starting point for hit-to-lead optimizations.


Subject(s)
Anthraquinones/chemistry , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , 5-Methylcytosine/chemistry , Binding Sites , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , Drug Evaluation, Preclinical/methods , HeLa Cells , Humans , Kinetics , Methylation , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Conformation , Structure-Activity Relationship , Transfection/methods , Ubiquitin-Protein Ligases
8.
Chemistry ; 24(52): 13850-13861, 2018 Sep 18.
Article in English | MEDLINE | ID: mdl-29989220

ABSTRACT

Nucleic acids are characterized by a variety of dynamically interconverting structures that play a major role in transcriptional and translational regulation as well as recombination and repair. To monitor these interconversions, Förster resonance energy transfer (FRET)-based techniques can be used, but require two fluorophores that are typically large and can alter the DNA/RNA structure and protein binding. Additionally, events that do not alter the donor/acceptor distance and/or angular relationship are frequently left undetected. A more benign approach relies on fluorescent nucleobases that can substitute their native counterparts with minimal perturbation, such as the recently developed 2-thienyl-3-hydroxychromone (3HCnt) and thienoguanosine (th G). To demonstrate the potency of 3HCnt and th G in deciphering interconversion mechanisms, we used the conversion of the (-)DNA copy of the HIV-1 primer binding site (-)PBS stem-loop into (+)/(-)PBS duplex, as a model system. When incorporated into the (-)PBS loop, the two probes were found to be highly sensitive to the individual steps both in the absence and the presence of a nucleic acid chaperone, providing the first complete mechanistic description of this critical process in HIV-1 replication. The combination of the two distinct probes appears to be instrumental for characterizing structural transitions of nucleic acids under various stimuli.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Nucleic Acids/chemistry , Nucleosides/chemistry , Binding Sites , Fluorescent Dyes/chemistry , Kinetics , Nucleic Acid Conformation , Protein Binding , Protein Conformation , Proteins/chemistry , Thermodynamics
9.
J Am Chem Soc ; 139(6): 2520-2528, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28112929

ABSTRACT

DNA methylation patterns, which are critical for gene expression, are replicated by DNA methyltransferase 1 (DNMT1) and ubiquitin-like containing PHD and RING finger domains 1 (UHRF1) proteins. This replication is initiated by the recognition of hemimethylated CpG sites and further flipping of methylated cytosines (mC) by the Set and Ring Associated (SRA) domain of UHRF1. Although crystallography has shed light on the mechanism of mC flipping by SRA, tools are required to monitor in real time how SRA reads DNA and flips the modified nucleobase. To accomplish this aim, we have utilized two distinct fluorescent nucleobase surrogates, 2-thienyl-3-hydroxychromone nucleoside (3HCnt) and thienoguanosine (thG), incorporated at different positions into hemimethylated (HM) and nonmethylated (NM) DNA duplexes. Large fluorescence changes were associated with mC flipping in HM duplexes, showing the outstanding sensitivity of both nucleobase surrogates to the small structural changes accompanying base flipping. Importantly, the nucleobase surrogates marginally affected the structure of the duplex and its affinity for SRA at positions where they were responsive to base flipping, illustrating their promise as nonperturbing probes for monitoring such events. Stopped-flow studies using these two distinct tools revealed the fast kinetics of SRA binding and sliding to NM duplexes, consistent with its reader role. In contrast, the kinetics of mC flipping was found to be much slower in HM duplexes, substantially increasing the lifetime of CpG-bound UHRF1, and thus the probability of recruiting DNMT1 to faithfully duplicate the DNA methylation profile. The fluorescence-based approach using these two different fluorescent nucleoside surrogates advances the mechanistic understanding of the UHRF1/DNMT1 tandem and the development of assays for the identification of base flipping inhibitors.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cytosine/metabolism , DNA/metabolism , Thermodynamics , CCAAT-Enhancer-Binding Proteins/chemistry , Cytosine/chemistry , DNA/chemistry , DNA Methylation , DNA Replication , Fluorescence , Humans , Kinetics , Molecular Structure , Ubiquitin-Protein Ligases
10.
J Biol Chem ; 291(10): 5116-27, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26792864

ABSTRACT

The low density lipoprotein receptor-related protein 1 (LRP1) is a ubiquitously expressed cell surface receptor that protects from intracellular cholesterol accumulation. However, the underlying mechanisms are unknown. Here we show that the extracellular (α) chain of LRP1 mediates TGFß-induced enhancement of Wnt5a, which limits intracellular cholesterol accumulation by inhibiting cholesterol biosynthesis and by promoting cholesterol export. Moreover, we demonstrate that the cytoplasmic (ß) chain of LRP1 suffices to limit cholesterol accumulation in LRP1(-/-) cells. Through binding of Erk2 to the second of its carboxyl-terminal NPXY motifs, LRP1 ß-chain positively regulates the expression of ATP binding cassette transporter A1 (ABCA1) and of neutral cholesterol ester hydrolase (NCEH1). These results highlight the unexpected functions of LRP1 and the canonical Wnt5a pathway and new therapeutic potential in cholesterol-associated disorders including cardiovascular diseases.


Subject(s)
Cholesterol/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway , ATP Binding Cassette Transporter 1/metabolism , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, LDL/chemistry , Receptors, LDL/genetics , Sterol Esterase/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein
11.
J Mol Biol ; 427(6 Pt B): 1480-1494, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25644662

ABSTRACT

The Pr55 Gag of human immunodeficiency virus type 1 orchestrates viral particle assembly in producer cells, which requires the genomic RNA and a lipid membrane as scaffolding platforms. The nucleocapsid (NC) domain with its two invariant CCHC zinc fingers flanked by unfolded basic sequences is thought to direct genomic RNA selection, dimerization and packaging during virus assembly. To further investigate the role of NC domain, we analyzed the assembly of Gag with deletions in the NC domain in parallel with that of wild-type Gag using fluorescence lifetime imaging microscopy combined with Förster resonance energy transfer in HeLa cells. We found that, upon binding to nucleic acids, the NC domain promotes the formation of compact Gag oligomers in the cytoplasm. Moreover, the intracellular distribution of the population of oligomers further suggests that oligomers progressively assemble during their trafficking toward the plasma membrane (PM), but with no dramatic changes in their compact arrangement. This ultimately results in the accumulation at the PM of closely packed Gag oligomers that likely arrange in hexameric lattices, as revealed by the perfect match between the experimental Förster resonance energy transfer value and the one calculated from the structural model of Gag in immature viruses. The distal finger and flanking basic sequences, but not the proximal finger, appear to be essential for Gag oligomer compaction and membrane binding. Moreover, the full NC domain was found to be instrumental in the kinetics of Gag oligomerization and intracellular trafficking. These findings further highlight the key roles played by the NC domain in virus assembly.


Subject(s)
Cell Membrane/metabolism , HIV Infections/metabolism , Microscopy, Fluorescence , Zinc Fingers/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , Cytoplasm/metabolism , Fluorescence Resonance Energy Transfer , HIV Infections/virology , HIV-1/physiology , Humans , Mutation/genetics , Nucleocapsid , Protein Binding , Protein Multimerization , Protein Transport , RNA, Viral/genetics , Virus Assembly , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics
12.
ACS Chem Biol ; 9(9): 1950-5, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-24988251

ABSTRACT

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.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Nucleocapsid Proteins/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Calorimetry/methods , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , HIV-1/chemistry , HIV-1/drug effects , HeLa Cells/drug effects , HeLa Cells/virology , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Docking Simulation , Nucleocapsid Proteins/metabolism , Structure-Activity Relationship , Thiazoles/chemistry
13.
Lab Chip ; 14(10): 1767-74, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24683603

ABSTRACT

We developed a new experimental approach combining Time-Resolved Fluorescence (TRF) spectroscopy and Droplet Microfluidics (DµF) to investigate the relaxation dynamics of structurally heterogeneous biomolecular systems. Here DµF was used to produce with minimal material consumption an out-of-equilibrium, fluorescently labeled biomolecular complex by rapid mixing within the droplets. TRF detection was implemented with a streak camera to monitor the time evolution of the structural heterogeneity of the complex along its relaxation towards equilibrium while it propagates inside the microfluidic channel. The approach was validated by investigating the fluorescence decay kinetics of a model interacting system of bovine serum albumin and Patent Blue V. Fluorescence decay kinetics are acquired with very good signal-to-noise ratio and allow for global, multicomponent fluorescence decay analysis, evidencing heterogeneous structural relaxation over several 100 ms.


Subject(s)
Fluorescent Dyes/chemistry , Microfluidic Analytical Techniques/methods , Rosaniline Dyes/chemistry , Serum Albumin, Bovine/chemistry , Spectrometry, Fluorescence , Animals , Cattle , Kinetics , Microfluidic Analytical Techniques/instrumentation , Oils/chemistry , Serum Albumin, Bovine/metabolism , Signal-To-Noise Ratio , Time Factors , Water/chemistry
14.
Nucleic Acids Res ; 42(2): 1065-78, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24153111

ABSTRACT

The HIV-1 transactivator of transcription (Tat) protein is thought to stimulate reverse transcription (RTion). The Tat protein and, more specifically, its (44-61) domain were recently shown to promote the annealing of complementary DNA sequences representing the HIV-1 transactivation response element TAR, named dTAR and cTAR, that plays a key role in RTion. Moreover, the kinetic mechanism of the basic Tat(44-61) peptide in this annealing further revealed that this peptide constitutes a representative nucleic acid annealer. To further understand the structure-activity relationships of this highly conserved domain, we investigated by electrophoresis and fluorescence approaches the binding and annealing properties of various Tat(44-61) mutants. Our data showed that the Tyr47 and basic residues of the Tat(44-61) domain were instrumental for binding to cTAR through stacking and electrostatic interactions, respectively, and promoting its annealing with dTAR. Furthermore, the annealing efficiency of the mutants clearly correlates with their ability to rapidly associate and dissociate the complementary oligonucleotides and to promote RTion. Thus, transient and dynamic nucleic acid interactions likely constitute a key mechanistic component of annealers and the role of Tat in the late steps of RTion. Finally, our data suggest that Lys50 and Lys51 acetylation regulates Tat activity in RTion.


Subject(s)
HIV Long Terminal Repeat , HIV-1 , Reverse Transcription , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Base Sequence , Molecular Sequence Data , Nucleic Acid Conformation , Oligonucleotides/chemistry , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , tat Gene Products, Human Immunodeficiency Virus/chemistry
15.
J Biol Chem ; 288(36): 25895-25907, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23843463

ABSTRACT

The antithrombin (AT) binding properties of heparin and low molecular weight heparins are strongly associated to the presence of the pentasaccharide sequence AGA*IA (A(NAc,6S)-GlcUA-A(NS,3,6S)-I(2S)-A(NS,6S)). By using the highly chemoselective depolymerization to prepare new ultra low molecular weight heparin and coupling it with the original separation techniques, it was possible to isolate a polysaccharide with a biosynthetically unexpected structure and excellent antithrombotic properties. It consisted of a dodecasaccharide containing an unsaturated uronate unit at the nonreducing end and two contiguous AT-binding sequences separated by a nonsulfated iduronate residue. This novel oligosaccharide was characterized by NMR spectroscopy, and its binding with AT was determined by fluorescence titration, NMR, and LC-MS. The dodecasaccharide displayed a significantly increased anti-FXa activity compared with those of the pentasaccharide, fondaparinux, and low molecular weight heparin enoxaparin.


Subject(s)
Factor Xa/chemistry , Fibrinolytic Agents , Oligosaccharides , Carbohydrate Sequence , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/chemistry , Heparin , Humans , Magnetic Resonance Spectroscopy , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry
16.
Biochem J ; 449(2): 343-51, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23083208

ABSTRACT

The 3-O-sulfation of N-sulfated glucosamine is the last event in the biosynthesis of heparin/heparan sulfate, giving rise to the antithrombin-binding pentasaccharide sequence AGA*IA, which is largely associated with the antithrombotic activity of these molecules. The aim of the present study was the structural and biochemical characterization of a previously unreported AGA*IA*-containing octasaccharide isolated from the very-low-molecular-mass heparin semuloparin, in which both glucosamine residues of the pentasaccharide moiety located at the non-reducing end bear 3-O-sulfate groups. Two-dimensional and STD (saturation transfer difference) NMR experiments clearly confirmed its structure and identified its ligand epitope binding to antithrombin. The molecular conformation of the octasaccharide-antithrombin complex has been determined by NMR experiments and docking/energy minimization. The presence of the second 3-O-sulfated glucosamine in the octasaccharide induced more than one order of magnitude increase in affinity to antithrombin compared to the pentasaccharide AGA*IA.


Subject(s)
Antithrombins/chemistry , Glucosamine/chemistry , Heparin/chemistry , Oligosaccharides/chemistry , Antithrombins/metabolism , Carbohydrate Sequence , Glucosamine/metabolism , Heparin/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Molecular Structure , Oligosaccharides/metabolism , Protein Binding , Protein Conformation , Sulfates/chemistry , Sulfates/metabolism , Temperature
17.
Nat Commun ; 3: 1077, 2012.
Article in English | MEDLINE | ID: mdl-23011131

ABSTRACT

Vascular calcification is a hallmark of advanced atherosclerosis. Here we show that deletion of the nuclear receptor PPARγ in vascular smooth muscle cells of low density lipoprotein receptor (LDLr)-deficient mice fed an atherogenic diet high in cholesterol, accelerates vascular calcification with chondrogenic metaplasia within the lesions. Vascular calcification in the absence of PPARγ requires expression of the transmembrane receptor LDLr-related protein-1 in vascular smooth muscle cells. LDLr-related protein-1 promotes a previously unknown Wnt5a-dependent prochondrogenic pathway. We show that PPARγ protects against vascular calcification by inducing the expression of secreted frizzled-related protein-2, which functions as a Wnt5a antagonist. Targeting this signalling pathway may have clinical implications in the context of common complications of atherosclerosis, including coronary artery calcification and valvular sclerosis.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , PPAR gamma/metabolism , Vascular Calcification/metabolism , Animals , Humans , Immunoblotting , Immunoprecipitation , In Situ Hybridization , In Vitro Techniques , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Myocytes, Smooth Muscle/drug effects , PPAR gamma/agonists , PPAR gamma/genetics , Rosiglitazone , Thiazolidinediones/pharmacology , Vascular Calcification/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein
18.
Virus Res ; 169(2): 349-60, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22743066

ABSTRACT

RNA chaperones are proteins able to rearrange nucleic acid structures towards their most stable conformations. In retroviruses, the reverse transcription of the viral RNA requires multiple and complex nucleic acid rearrangements that need to be chaperoned. HIV-1 has evolved different viral-encoded proteins with chaperone activity, notably Tat and the well described nucleocapsid protein NCp7. We propose here an overview of the recent reports that examine and compare the nucleic acid chaperone properties of Tat and NCp7 during reverse transcription to illustrate the variety of mechanisms of action of the nucleic acid chaperone proteins.


Subject(s)
HIV-1/physiology , Molecular Chaperones/metabolism , RNA, Transfer, Lys/metabolism , RNA, Viral/metabolism , Reverse Transcription , gag Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism
19.
Antimicrob Agents Chemother ; 56(2): 1010-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22083480

ABSTRACT

Since currently available therapies against HIV/AIDS still show important drawbacks, the development of novel anti-HIV treatments is a key issue. We recently characterized methylated oligoribonucleotides (mONs) that extensively inhibit HIV-1 replication in primary T cells at nanomolar concentrations. The mONs were shown to target both HIV-1 reverse transcriptase (RT) and the nucleocapsid protein (NC), which is an essential partner of RT during viral DNA synthesis. To further understand the mechanism of such mONs, we studied by isothermal titration calorimetry and fluorescence-based techniques their NC binding properties and ability to inhibit the nucleic acid chaperone properties of NC. Notably, we investigated the ability of mONs to inhibit the NC-induced destabilization of the HIV-1 cTAR (complementary DNA sequence to TAR [transactivation response element]) stem-loop and the NC-promoted cTAR annealing to its complementary sequence, required at the early stage of HIV-1 viral DNA synthesis. Moreover, we compared the activity of the mONs to that of a number of modified and nonmodified oligonucleotides. Results show that the mONs inhibit NC by a competitive mechanism whereby the mONs tightly bind the NC peptide, mainly through nonelectrostatic interactions with the hydrophobic platform at the top of the NC zinc fingers. Taken together, these results favor the notion that the mONs impair the process of the RT-directed viral DNA synthesis by sequestering NC molecules, thus preventing the chaperoning of viral DNA synthesis by NC. These findings contribute to the understanding of the molecular basis for NC inhibition by mONs, which could be used for the rational design of antiretroviral compounds targeting HIV-1 NC protein.


Subject(s)
HIV-1/metabolism , Molecular Chaperones/antagonists & inhibitors , Nucleocapsid Proteins/antagonists & inhibitors , Oligoribonucleotides/pharmacology , Amino Acid Sequence , Base Sequence , DNA, Viral/biosynthesis , HIV Reverse Transcriptase/antagonists & inhibitors , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism
20.
Biochimie ; 93(10): 1647-58, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704113

ABSTRACT

Addition of Vpr C-terminus to various cell types provokes cell apoptosis. This property was recently shown useful to develop inhibitors of cell proliferation. In that context, we investigated the cellular uptake of rhodamine- and fluorescein-labeled Vpr(52-96) peptides to understand the mechanism of Vpr C-terminus entry into cells. Dynamic light scattering data indicated that this peptide spontaneously formed polydispersed aggregates in cell culture medium. The fluorescently labeled Vpr(52-96) peptide was efficiently internalized, appearing either as large fluorescent patches in the cytoplasm or in a more diffuse form throughout the cell. Using isothermal titration calorimetry, we demonstrated that Vpr(52-96) can tightly associate with heparin, a glycosaminoglycan analog of heparan sulphate, suggesting a central role of the ubiquitous cell surface-associated heparan sulphate proteoglycans for the internalization of Vpr C-terminus. Fluorescently-labeled transferrin and methyl-ß-cyclodextrin showed that the Vpr C-terminus was mediated through clathrin- and caveolae/raft-dependent endocytosis. We found that Vpr C-terminus uptake was partly blocked at 4°C suggesting the importance of membrane fluidity for Vpr C-terminus entry. In fact, atomic force microscopy and liposome leakage further indicated that the Vpr peptide can destabilize and disrupt model membrane bilayers, suggesting that this mechanism may contribute to the passive entry of the peptide. Finally, using fluorescence lifetime imaging, we found that the Vpr(52-96) peptide was stable in cells for at least 48h, probably as a consequence of the poor accessibility of the peptide to proteolytic enzymes in aggregates.


Subject(s)
vpr Gene Products, Human Immunodeficiency Virus/metabolism , Calorimetry , Endocytosis , HeLa Cells , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Liposomes/chemistry , Liposomes/metabolism , Microscopy, Atomic Force , Microscopy, Confocal , Transferrin/chemistry , Transferrin/metabolism , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/metabolism , vpr Gene Products, Human Immunodeficiency Virus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...