Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochemistry (Mosc) ; 87(11): 1292-1300, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36509716

RESUMO

The process of unfolding of G-quadruplex structure in the RE31 DNA-aptamer and in its complex with thrombin under the action of the fluorescently labeled complementary oligonucleotides of varying length with formation of double-helix structures has been studied. It has been suggested that G-quadruplex unfolding involves formation of an intermediate complex with an oligonucleotide. Thermodynamic parameters and kinetics of unfolding of the free aptamer and its complex with thrombin differ. Extension of the oligonucleotide sequence complementary to G-quadruplex by two nucleotides to cover the so-called "hinge region" had little impact on the conformational transition of G-quadruplex of the free aptamer. However, a pronounced effect has been observed for the aptamer-protein complex. Most likely these differences could be explained by the thrombin-induced conformational transition of the aptamer involving the hinge region.


Assuntos
Aptâmeros de Nucleotídeos , Quadruplex G , Aptâmeros de Nucleotídeos/química , Trombina/metabolismo , Termodinâmica , Cinética
2.
Nucleic Acids Res ; 46(22): 12177-12185, 2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30357392

RESUMO

Despite aptamers are very promising alternative to antibodies, very few of them are under clinical trials or are used as drugs. Among them, NU172 is currently in Phase II as anticoagulant in heart disease treatments. It inhibits thrombin activity much more effectively than TBA, the best-known thrombin binding aptamer. The crystal structure of thrombin-NU172 complex reveals a bimodular duplex/quadruplex architecture for the aptamer, which binds thrombin exosite I through a highly complementary surface involving all three loops of the G-quadruplex module. Although the duplex domain does not interact directly with thrombin, the features of the duplex/quadruplex junction and the solution data on two newly designed NU172 mutants indicate that the duplex moiety is important for the optimization of the protein-ligand interaction and for the inhibition of the enzyme activity. Our work discloses the structural features determining the inhibition of thrombin by NU172 and put the basis for the design of mutants with improved properties.


Assuntos
Aptâmeros de Nucleotídeos/química , Fibrinolíticos/química , Trombina/química , Motivos de Aminoácidos , Anticoagulantes/química , Dicroísmo Circular , Cristalografia por Raios X , Fibrinogênio/química , Quadruplex G , Humanos , Ligantes , Modelos Moleculares , Mutação , Oligonucleotídeos/química , Ligação Proteica , Conformação Proteica
3.
Anal Bioanal Chem ; 411(25): 6723-6732, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31396648

RESUMO

Nucleic acid (NA) aptamers bind to their targets with high affinity and selectivity. The three-dimensional (3D) structures of aptamers play a major role in these non-covalent interactions. Here, we use a four-step approach to determine a true 3D structure of aptamers in solution using small-angle X-ray scattering (SAXS) and molecular structure restoration (MSR). The approach consists of (i) acquiring SAXS experimental data of an aptamer in solution, (ii) building a spatial distribution of the molecule's electron density using SAXS results, (iii) constructing a 3D model of the aptamer from its nucleotide primary sequence and secondary structure, and (iv) comparing and refining the modeled 3D structures with the experimental SAXS model. In the proof-of-principle we analyzed the 3D structure of RE31 aptamer to thrombin in a native free state at different temperatures and validated it by circular dichroism (CD). The resulting 3D structure of RE31 has the most energetically favorable conformation and the same elements such as a B-form duplex, non-complementary region, and two G-quartets which were previously reported by X-ray diffraction (XRD) from a single crystal. More broadly, this study demonstrates the complementary approach for constructing and adjusting the 3D structures of aptamers, DNAzymes, and ribozymes in solution, and could supply new opportunities for developing functional nucleic acids. Graphical abstract.


Assuntos
Aptâmeros de Nucleotídeos/química , Algoritmos , Simulação por Computador , Quadruplex G , Modelos Moleculares , Conformação de Ácido Nucleico , Espalhamento a Baixo Ângulo , Difração de Raios X/métodos
4.
Nucleic Acids Res ; 44(2): 983-91, 2016 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-26673709

RESUMO

Mixed duplex/quadruplex oligonucleotides have attracted great interest as therapeutic targets as well as effective biomedical aptamers. In the case of thrombin-binding aptamer (TBA), the addition of a duplex motif to the G-quadruplex module improves the aptamer resistance to biodegradation and the affinity for thrombin. In particular, the mixed oligonucleotide RE31 is significantly more effective than TBA in anticoagulation experiments and shows a slower disappearance rate in human plasma and blood. In the crystal structure of the complex with thrombin, RE31 adopts an elongated structure in which the duplex and quadruplex regions are perfectly stacked on top of each other, firmly connected by a well-structured junction. The lock-and-key shape complementarity between the TT loops of the G-quadruplex and the protein exosite I gives rise to the basic interaction that stabilizes the complex. However, our data suggest that the duplex motif may have an active role in determining the greater anti-thrombin activity in biological fluids with respect to TBA. This work gives new information on mixed oligonucleotides and highlights the importance of structural data on duplex/quadruplex junctions, which appear to be varied, unpredictable, and fundamental in determining the aptamer functional properties.


Assuntos
Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/metabolismo , Trombina/química , Aptâmeros de Nucleotídeos/farmacologia , Cristalografia por Raios X , Quadruplex G , Ligação de Hidrogênio , Modelos Moleculares , Conformação de Ácido Nucleico , Trombina/antagonistas & inibidores , Trombina/metabolismo
6.
Int J Biol Macromol ; 107(Pt B): 1697-1705, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29024684

RESUMO

Recently, mixed duplex/quadruplex oligonucleotides have attracted great interest for use as biomedical aptamers. In the case of anti-thrombin aptamers, the addition of duplex-forming sequences to a G-quadruplex module identical or very similar to the best-known G-quadruplex of the Thrombin Binding Aptamer (HD1) results in new or improved biological properties, such as higher activity or different recognition properties with respect to HD1. Remarkably, this bimodular fold was hypothesized, based on its sequence, for the only anti-thrombin aptamer in advanced clinical trial, NU172. Whereas cation modulation of G-quadruplex conformation and stability is well characterized, only few data from similar analysis on duplex/quadruplex oligonucleotides exist. Here we have performed a characterization of structure and stability of four different duplex/quadruplex anti-thrombin aptamers, including NU172, in the presence of different cations and in physiological-mimicking conditions in comparison to HD1, by means of spectroscopic techniques (UV and circular dichroism) and differential scanning calorimetry. Our data show a strong reciprocal influence of each domain on the stability of the other and in particular suggest a stabilizing effect of the duplex region in the presence of solutions mimicking the physiological conditions, strengthening the idea that bimodular aptamers present better therapeutic potentialities than those containing a single G-quadruplex domain.


Assuntos
Antitrombinas/farmacologia , Aptâmeros de Nucleotídeos/farmacologia , Quadruplex G , Oligonucleotídeos/química , Sequência de Bases , Varredura Diferencial de Calorimetria , Dicroísmo Circular , Desnaturação de Ácido Nucleico , Temperatura , Raios Ultravioleta
7.
FEBS Lett ; 580(25): 5858-62, 2006 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-17027976

RESUMO

In E. coli, S7 initiates 30S ribosome assembly by binding to 16S rRNA. It also regulates translation of the S12 and S7 cistrons of the 'streptomycin' operon transcript by binding to the S12-S7 intercistronic region. Here, we describe the contacts of N-terminally His(6)-tagged S7 with this region as mapped by UV-induced cross-linking. The cross-links are located at U(-34), U(-35), quite distant from the start codons of the two cistrons. In order to explain the mechanism of translational repression of S12-S7, we consider a possible conformational rearrangement of the intercistronic RNA structure induced by S7 binding.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Bacterianos , Proteínas Ribossômicas/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Mapeamento Cromossômico , Proteínas de Escherichia coli/genética , Genes , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Óperon , Biossíntese de Proteínas , RNA Bacteriano/química , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Ribossômicas/genética , Estreptomicina/biossíntese
8.
Nucleic Acids Res ; 32(8): 2594-7, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15141029

RESUMO

Tetracycline blocks stable binding of aminoacyl-tRNA to the bacterial ribosomal A-site. Various tetracycline binding sites have been identified in crystals of the 30S ribosomal small subunit of Thermus thermophilus. Here we describe a direct photo- affinity modification of the ribosomal small subunits of Escherichia coli with 7-[3H]-tetracycline. To select for specific interactions, an excess of the 30S subunits over tetracycline has been used. Primer extension analysis of the 16S rRNA revealed two sites of the modifications: C936 and C948. Considering available data on tetracycline interactions with the prokaryotic 30S subunits, including the presented data (E.coli), X-ray data (T.thermophilus) and genetic data (Helicobacter pylori, E.coli), a second high affinity tetracycline binding site is proposed within the 3'-major domain of the 16S rRNA, in addition to the A-site related tetracycline binding site.


Assuntos
Antibacterianos/metabolismo , Escherichia coli/metabolismo , Inibidores da Síntese de Proteínas/metabolismo , RNA Ribossômico 16S/metabolismo , Ribossomos/metabolismo , Tetraciclina/metabolismo , Antibacterianos/química , Sítios de Ligação , Escherichia coli/genética , Modelos Moleculares , Inibidores da Síntese de Proteínas/química , RNA Ribossômico 16S/química , Ribossomos/química , Tetraciclina/química
9.
Blood Coagul Fibrinolysis ; 25(1): 39-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24247320

RESUMO

Investigation of inhibitory effect of two single-stranded DNA thrombin-inhibiting aptamers (15TBA and 31TBA) on fibrin polymerization in fibrinogen solutions and comparison of anticoagulant properties of these aptamers by a new global coagulation test of thrombodynamics. Measurement of aptamers' functional stability in human plasma and blood in vitro in order to investigate the involvement of 3'-exonuclease in fast decrease of aptamers' functional activity in vivo. Thrombin inhibition activity was measured in a buffer system in vitro as effects of aptamers on fibrin polymerization. Anticoagulant activity was investigated by measuring the spatial clot growth rate in the presence of aptamers. The stability of aptamers during incubation in human plasma was investigated in vitro by measuring activated partial thromboplastin time. Both aptamers dose-dependently inhibit fibrin polymerization in a buffer solution (IC50=10 nm for 15TBA and 3 nm for 31TBA) and are effective anticoagulants in human plasma (IC50 for spatial clot growth rate decreasing are 9.5 µmol/l and 4.0 µmol/l for 15TBA and 31TBA, correspondingly). Both aptamers remain stable in plasma or whole blood in vitro for at least 4 h. It was shown that 31TBA was 2-3 times more effective than 15TBA. Both aptamers were stable in human plasma and whole blood in vitro. So, the 3'-exonuclease could not be the reason for fast decrease of aptamers' functional activity in vivo. The main role in the removal of oligonucleotides from the circulation is played obviously by the liver.


Assuntos
Anticoagulantes/farmacologia , Aptâmeros de Nucleotídeos/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , DNA de Cadeia Simples/farmacologia , Trombina/antagonistas & inibidores , Anticoagulantes/química , Aptâmeros de Nucleotídeos/química , DNA de Cadeia Simples/química , Estabilidade de Medicamentos , Fibrina/química , Fibrina/metabolismo , Fibrinogênio/química , Fibrinogênio/metabolismo , Humanos
10.
J Chem Theory Comput ; 6(10): 3003-14, 2010 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26616765

RESUMO

The thrombin-binding aptamer (15-TBA) is a 15-mer DNA oligonucleotide with sequence d(GGTTGGTGTGGTTGG). 15-TBA folds into a quadruplex DNA (G-DNA) structure with two planar G-quartets connected by three single-stranded loops. The arrangement of the 15-TBA-thrombin complex is unclear, particularly with respect to the precise 15-TBA residues that interact with the thrombin structure. Our present understanding suggests either the 15-TBA single stranded loops containing sequential thymidines (TT) or alternatively a single-stranded loop, containing a guanine flanked by 2 thymidines (TGT), physically associates with thrombin protein. In the present study, the explicit solvent molecular dynamics (MD) simulation method was utilized to further analyze the 15-TBA-thrombin three-dimensional structure. Functional annotation of the loop residues was made with long simulations in the parmbsc0 force field. In total, the elapsed time of simulations carried out in this study exceeds 12 microseconds, substantially surpassing previous G-DNA simulation reports. Our simulations suggest that the TGT-loop function is to stabilize the structure of the aptamer, while the TT-loops participate in direct binding to thrombin. The findings of the present report advance our understanding of the molecular structure of the 15-TBA-thrombin structure further enabling the construction of biosensors for aptamer bases and the development of anticoagulant agents.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA