Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Antiviral Res ; 208: 105458, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36336176

RESUMO

Severe acute respiratory syndrome coronaviruses 1 and 2 (SARS-CoV-1 and SARS-CoV-2) pose a threat to global public health. The 3C-like main protease (Mpro), which presents structural similarity with the active site domain of enterovirus 3C protease, is one of the best-characterized drug targets of these viruses. Here we studied the antiviral activity of the orally bioavailable enterovirus protease inhibitor AG7404 against SARS-CoV-1 and SARS-CoV-2 from a structural, biochemical, and cellular perspective, comparing it with the related molecule rupintrivir (AG7800). Crystallographic structures of AG7404 in complex with SARS-CoV-1 Mpro and SARS-CoV-2 Mpro and of rupintrivir in complex with SARS-CoV-2 Mpro were solved, revealing that all protein residues interacting with the inhibitors are conserved between the two proteins. A detailed analysis of protein-inhibitor interactions indicates that AG7404 has a better fit to the active site of the target protease than rupintrivir. This observation was further confirmed by biochemical FRET assays showing IC50 values of 47 µM and 101 µM for AG7404 and rupintrivir, respectively, in the case of SARS-CoV-2 Mpro. Equivalent IC50 values for SARS-CoV-1 also revealed greater inhibitory capacity of AG7404, with a value of 29 µM vs. 66 µM for rupintrivir. Finally, the antiviral activity of the two inhibitors against SARS-CoV-2 was confirmed in a human cell culture model of SARS-CoV-2 infection, although rupintrivir showed a higher potency and selectivity index in this assay.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Humanos , Antivirais/química , Cisteína Endopeptidases/metabolismo , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química , Simulação de Acoplamento Molecular
2.
Nat Commun ; 10(1): 3891, 2019 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-31467275

RESUMO

Herpesviridae is a vast family of enveloped DNA viruses that includes eight distinct human pathogens, responsible for diseases that range from almost asymptomatic to severe and life-threatening. Epstein-Barr virus infects B-cells and epithelial cells, causing infectious mononucleosis, as well as a number of cancers. Epstein-Barr infection cannot be cured since neither vaccine nor antiviral drug treatments are available. All herpesviruses contain a linear double-stranded DNA genome, enclosed within an icosahedral capsid. Viral portal protein plays a key role in the procapsid assembly and DNA packaging. The portal is the entrance and exit pore for the viral genome, making it an attractive pharmacological target for the development of new antivirals. Here we present the atomic structure of the portal protein of Epstein-Barr virus, solved by cryo-electron microscopy at 3.5 Å resolution. The detailed architecture of this protein suggests that it plays a functional role in DNA retention during packaging.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Herpesvirus Humano 4/ultraestrutura , Proteínas Virais/ultraestrutura , Montagem de Vírus , Capsídeo/ultraestrutura , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Empacotamento do DNA , DNA Viral/genética , Genoma Viral , Herpesvirus Humano 4/genética , Humanos , Modelos Moleculares , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/ultraestrutura , Proteínas Virais/genética , Vírion/ultraestrutura
3.
RNA ; 25(5): 607-619, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30737359

RESUMO

Adenosine deaminase acting on transfer RNA (ADAT) is an essential eukaryotic enzyme that catalyzes the deamination of adenosine to inosine at the first position of tRNA anticodons. Mammalian ADATs modify eight different tRNAs, having increased their substrate range from a bacterial ancestor that likely deaminated exclusively tRNAArg Here we investigate the recognition mechanisms of tRNAArg and tRNAAla by human ADAT to shed light on the process of substrate expansion that took place during the evolution of the enzyme. We show that tRNA recognition by human ADAT does not depend on conserved identity elements, but on the overall structural features of tRNA. We find that ancestral-like interactions are conserved for tRNAArg, while eukaryote-specific substrates use alternative mechanisms. These recognition studies show that human ADAT can be inhibited by tRNA fragments in vitro, including naturally occurring fragments involved in important regulatory pathways.


Assuntos
Adenosina Desaminase/metabolismo , Anticódon/química , RNA de Transferência de Alanina/química , RNA de Transferência de Arginina/química , Adenosina/metabolismo , Adenosina Desaminase/genética , Anticódon/genética , Anticódon/metabolismo , Sequência de Bases , Desaminação , Evolução Molecular , Expressão Gênica , Humanos , Inosina/metabolismo , Conformação de Ácido Nucleico , RNA de Transferência de Alanina/genética , RNA de Transferência de Alanina/metabolismo , RNA de Transferência de Arginina/genética , RNA de Transferência de Arginina/metabolismo , Alinhamento de Sequência , Especificidade por Substrato
4.
PLoS One ; 12(10): e0186286, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29045454

RESUMO

Sulfur trafficking in living organisms relies on transpersulfuration reactions consisting in the enzyme-catalyzed transfer of S atoms via activated persulfidic S across protein-protein interfaces. The recent elucidation of the mechanistic basis for transpersulfuration in the CsdA-CsdE model system has paved the way for a better understanding of its role under oxidative stress. Herein we present the crystal structure of the oxidized, inactivated CsdE dimer at 2.4 Å resolution. The structure sheds light into the activation of the Cys61 nucleophile on its way from a solvent-secluded position in free CsdE to a fully extended conformation in the persulfurated CsdA-CsdE complex. Molecular dynamics simulations of available CsdE structures allow to delineate the sequence of conformational changes underwent by CsdE and to pinpoint the key role played by the deprotonation of the Cys61 thiol. The low-energy subunit orientation in the disulfide-bridged CsdE dimer demonstrates the likely physiologic relevance of this oxidative dead-end form of CsdE, suggesting that CsdE could act as a redox sensor in vivo.


Assuntos
Liases de Carbono-Enxofre/química , RNA Helicases DEAD-box/química , Proteínas de Escherichia coli/química , Conformação Proteica , Enxofre/química , Liases de Carbono-Enxofre/genética , Cristalografia por Raios X , RNA Helicases DEAD-box/genética , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Simulação de Dinâmica Molecular , Estresse Oxidativo/genética , Domínios e Motivos de Interação entre Proteínas/genética , Multimerização Proteica , Enxofre/metabolismo
5.
Sci Rep ; 7: 39680, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-28051169

RESUMO

Variolin B is a rare marine alkaloid that showed promising anti-cancer activity soon after its isolation. It acts as a cyclin-dependent kinase inhibitor, although the precise mechanism through which it exerts the cytotoxic effects is still unknown. The crystal structure of a variolin B bound to a DNA forming a pseudo-Holliday junction shows that this compound can also contribute, through intercalative binding, to either the formation or stabilization of multi-stranded DNA forms.


Assuntos
Antineoplásicos/química , Compostos Aza/química , DNA/química , Substâncias Intercalantes/química , Pirimidinas/química , Cristalografia por Raios X , Modelos Moleculares , Relação Estrutura-Atividade
6.
PLoS One ; 10(4): e0118606, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25897750

RESUMO

Cyclic N6-threonylcarbamoyladenosine ('cyclic t6A', ct(6)A) is a non-thiolated hypermodification found in transfer RNAs (tRNAs) in bacteria, protists, fungi and plants. In bacteria and yeast cells ct(6)A has been shown to enhance translation fidelity and efficiency of ANN codons by improving the faithful discrimination of aminoacylated tRNAs by the ribosome. To further the understanding of ct(6)A biology we have determined the high-resolution crystal structures of CsdL/TcdA in complex with AMP and ATP, an E1-like activating enzyme from Escherichia coli, which catalyzes the ATP-dependent dehydration of t6A to form ct(6)A. CsdL/TcdA is a dimer whose structural integrity and dimer interface depend critically on strongly bound K+ and Na+ cations. By using biochemical assays and small-angle X-ray scattering we show that CsdL/TcdA can associate with tRNA with a 1:1 stoichiometry and with the proper position and orientation for the cyclization of t6A. Furthermore, we show by nuclear magnetic resonance that CsdL/TcdA engages in transient interactions with CsdA and CsdE, which, in the latter case, involve catalytically important residues. These short-lived interactions may underpin the precise channeling of sulfur atoms from cysteine to CsdL/TcdA as previously characterized. In summary, the combination of structural, biophysical and biochemical methods applied to CsdL/TcdA has afforded a more thorough understanding of how the structure of this E1-like enzyme has been fine tuned to accomplish ct(6)A synthesis on tRNAs while providing support for the notion that CsdA and CsdE are able to functionally interact with CsdL/TcdA.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , RNA de Transferência/metabolismo , Enzimas Ativadoras de Ubiquitina/química , Enzimas Ativadoras de Ubiquitina/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Escherichia coli/química , Modelos Moleculares , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Conformação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
7.
Biochim Biophys Acta ; 1824(2): 339-49, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22138634

RESUMO

The joint substitution of three active-site residues in Escherichia coli (L)-aspartate aminotransferase increases the ratio of l-cysteine sulfinate desulfinase to transaminase activity 10(5)-fold. This change in reaction specificity results from combining a tyrosine-shift double mutation (Y214Q/R280Y) with a non-conservative substitution of a substrate-binding residue (I33Q). Tyr214 hydrogen bonds with O3 of the cofactor and is close to Arg374 which binds the α-carboxylate group of the substrate; Arg280 interacts with the distal carboxylate group of the substrate; and Ile33 is part of the hydrophobic patch near the entrance to the active site, presumably participating in the domain closure essential for the transamination reaction. In the triple-mutant enzyme, k(cat)' for desulfination of l-cysteine sulfinate increased to 0.5s(-1) (from 0.05s(-1) in wild-type enzyme), whereas k(cat)' for transamination of the same substrate was reduced from 510s(-1) to 0.05s(-1). Similarly, k(cat)' for ß-decarboxylation of l-aspartate increased from<0.0001s(-1) to 0.07s(-1), whereas k(cat)' for transamination was reduced from 530s(-1) to 0.13s(-1). l-Aspartate aminotransferase had thus been converted into an l-cysteine sulfinate desulfinase that catalyzes transamination and l-aspartate ß-decarboxylation as side reactions. The X-ray structures of the engineered l-cysteine sulfinate desulfinase in its pyridoxal-5'-phosphate and pyridoxamine-5'-phosphate form or liganded with a covalent coenzyme-substrate adduct identified the subtle structural changes that suffice for generating desulfinase activity and concomitantly abolishing transaminase activity toward dicarboxylic amino acids. Apparently, the triple mutation impairs the domain closure thus favoring reprotonation of alternative acceptor sites in coenzyme-substrate intermediates by bulk water.


Assuntos
Aspartato Aminotransferases/química , Liases de Carbono-Enxofre/química , Liases de Carbono-Enxofre/genética , Domínio Catalítico/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Aminoácidos Dicarboxílicos/metabolismo , Aspartato Aminotransferases/genética , Aspartato Aminotransferases/metabolismo , Biocatálise , Liases de Carbono-Enxofre/metabolismo , Cristalografia por Raios X , Escherichia coli , Ligação de Hidrogênio , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Engenharia de Proteínas , Fosfato de Piridoxal/química , Fosfato de Piridoxal/metabolismo , Piridoxamina/análogos & derivados , Piridoxamina/química , Piridoxamina/metabolismo , Especificidade por Substrato
8.
J Virol ; 85(20): 10764-73, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21835784

RESUMO

Members of the Enterovirus genus of the Picornaviridae family are abundant, with common human pathogens that belong to the rhinovirus (HRV) and enterovirus (EV) species, including diverse echo-, coxsackie- and polioviruses. They cause a wide spectrum of clinical manifestations ranging from asymptomatic to severe diseases with neurological and/or cardiac manifestations. Pandemic outbreaks of EVs may be accompanied by meningitis and/or paralysis and can be fatal. However, no effective prophylaxis or antiviral treatment against most EVs is available. The EV RNA genome directs the synthesis of a single polyprotein that is autocatalytically processed into mature proteins at Gln↓Gly cleavage sites by the 3C protease (3C(pro)), which has narrow, conserved substrate specificity. These cleavages are essential for virus replication, making 3C(pro) an excellent target for antivirus drug development. In this study, we report the first determination of the crystal structure of 3C(pro) from an enterovirus B, EV-93, a recently identified pathogen, alone and in complex with the anti-HRV molecules compound 1 (AG7404) and rupintrivir (AG7088) at resolutions of 1.9, 1.3, and 1.5 Å, respectively. The EV-93 3C(pro) adopts a chymotrypsin-like fold with a canonically configured oxyanion hole and a substrate binding pocket similar to that of rhino-, coxsackie- and poliovirus 3C proteases. We show that compound 1 and rupintrivir are both active against EV-93 in infected cells and inhibit the proteolytic activity of EV-93 3C(pro) in vitro. These results provide a framework for further structure-guided optimization of the tested compounds to produce antiviral drugs against a broad range of EV species.


Assuntos
Antivirais/metabolismo , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Enterovirus/química , Enterovirus/enzimologia , Inibidores Enzimáticos/metabolismo , Isoxazóis/metabolismo , Pirrolidinonas/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteases Virais 3C , Antivirais/química , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/química , Humanos , Isoxazóis/química , Modelos Moleculares , Fenilalanina/análogos & derivados , Ligação Proteica , Estrutura Terciária de Proteína , Pirrolidinonas/química , Valina/análogos & derivados
9.
FEBS J ; 278(2): 273-81, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21134126

RESUMO

Mouse liver glutathione transferase P1-1 has three cysteine residues at positions 14, 47 and 169. We have constructed the single, double and triple cysteine to alanine mutants to define the behaviour of all three thiols. We confirm that C47 is the 'fast' thiol (pK 7.4), and define C169 as the alkaline reactive residue with a pK(a) of 8.6. Only a small proportion of C14 is reactive with 5,5'-dithiobis-(2-nitrobenoic acid) (DTNB) at pH 9 in the C47A/C169A double mutant. The native enzyme and the C169A mutant exhibited Michaelis-Menten kinetics, but all other thiol to alanine mutants exhibited sigmoidal kinetics to varying degrees. The C169A mutant exhibited 'ping pong' kinetics, consistent with a mechanism whereby liberation of a proton from a reduced enzyme-glutathione (GSH) complex to form an enzyme-GS(-) (unprotonated) complex is essentially irreversible. Intriguingly, similar behaviour has recently been reported for a mutant of the yeast prion Ure2p. This cooperative behaviour is 'mirrored' in the crystal structure of the C47A mutant, which binds the p-nitrobenzyl moiety of p-nitrobenzyglutathione in distinct orientations in the two crystallographic subunits. The asymmetry seen in this structure for product binding is associated with absence of a water molecule W0 in the standard wild-type conformation of product binding that is clearly identifiable in the new structure, which may represent a structural model for binding of incoming GSH prior to displacement of W0. Elimination of W0 as a hydroxonium ion may be the mechanism for the initial proton extrusion from the active site.


Assuntos
Biocatálise , Glutationa S-Transferase pi/química , Glutationa/química , Mutagênese Sítio-Dirigida , Prótons , Água/química , Substituição de Aminoácidos/fisiologia , Animais , Domínio Catalítico/fisiologia , Cristalografia por Raios X , Cisteína/química , Cisteína/genética , Dinitroclorobenzeno/química , Ácido Ditionitrobenzoico/química , Glutationa/análogos & derivados , Glutationa/metabolismo , Glutationa S-Transferase pi/antagonistas & inibidores , Glutationa S-Transferase pi/genética , Glutationa S-Transferase pi/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Camundongos , Modelos Químicos , Modelos Moleculares , Ligação Proteica/fisiologia , Conformação Proteica , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
Dalton Trans ; (3): 399-414, 2009 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19122895

RESUMO

In this paper, we review recent DNA-binding agents that are expected to influence the field of DNA-targeting. We restrict ourselves to binders for which the three-dimensional structure in complex with DNA or RNA has been determined by X-ray crystallography or NMR. Furthermore, we primarily focus on unprecedented ways of targeting peculiar DNA structures, such as junctions, quadruplexes, and duplex DNAs different from the B-form. Classical binding modes of small molecular weight compounds to DNA, i.e. groove binding, intercalation and covalent addition are discussed in those cases where the structures represent a novelty. In addition, we review 3D structures of triple-stranded DNA, of the so-called Peptide Nucleic Acids (PNAs), which are oligonucleotide bases linked by a polypeptide backbone, and of aptamers, which are DNA or RNA receptors that are designed combinatorially. A discussion on perspectives in the field of DNA-targeting and on sequence recognition is also provided.


Assuntos
DNA/química , Técnicas de Química Combinatória , Cristalografia por Raios X/métodos , DNA Cruciforme/química , Desenho de Fármacos , Substâncias Intercalantes/química , Ligantes , Espectroscopia de Ressonância Magnética/métodos , Modelos Químicos , Conformação Molecular , Conformação de Ácido Nucleico , Oligonucleotídeos/química , Ácidos Nucleicos Peptídicos/química , Fosfatos/química , RNA/química , Estereoisomerismo
11.
J Bacteriol ; 187(14): 4957-66, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15995211

RESUMO

The FucO protein, a member of the group III "iron-activated" dehydrogenases, catalyzes the interconversion between L-lactaldehyde and L-1,2-propanediol in Escherichia coli. The three-dimensional structure of FucO in a complex with NAD(+) was solved, and the presence of iron in the crystals was confirmed by X-ray fluorescence. The FucO structure presented here is the first structure for a member of the group III bacterial dehydrogenases shown experimentally to contain iron. FucO forms a dimer, in which each monomer folds into an alpha/beta dinucleotide-binding N-terminal domain and an all-alpha-helix C-terminal domain that are separated by a deep cleft. The dimer is formed by the swapping (between monomers) of the first chain of the beta-sheet. The binding site for Fe(2+) is located at the face of the cleft formed by the C-terminal domain, where the metal ion is tetrahedrally coordinated by three histidine residues (His200, His263, and His277) and an aspartate residue (Asp196). The glycine-rich turn formed by residues 96 to 98 and the following alpha-helix is part of the NAD(+) recognition locus common in dehydrogenases. Site-directed mutagenesis and enzyme kinetic assays were performed to assess the role of different residues in metal, cofactor, and substrate binding. In contrast to previous assumptions, the essential His267 residue does not interact with the metal ion. Asp39 appears to be the key residue for discriminating against NADP(+). Modeling L-1,2-propanediol in the active center resulted in a close approach of the C-1 hydroxyl of the substrate to C-4 of the nicotinamide ring, implying that there is a typical metal-dependent dehydrogenation catalytic mechanism.


Assuntos
Aldeídos/metabolismo , Escherichia coli/enzimologia , Ferro/farmacologia , Oxirredutases/química , Propilenoglicol/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Cristalografia por Raios X/métodos , Primers do DNA , Modelos Moleculares , Dados de Sequência Molecular , Oxirredutases/metabolismo , Conformação Proteica , Estrutura Secundária de Proteína , Especificidade por Substrato
12.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 7): 1009-12, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15983425

RESUMO

Ellipticine is a natural plant product that has been found to be a powerful anticancer drug. Although still unclear, its mechanism of action is considered to be mainly based on DNA intercalation and/or the inhibition of topoisomerase II. Many experimental data suggest an intercalation based on stacking interactions along the major base-pair axis, but alternative binding modes have been proposed, in particular for ellipticine derivatives. The 1.5 A resolution structure of ellipticine complexed to a 6 bp oligonucleotide unveils its mode of binding and enables a detailed analysis of the distorting effects of the drug on the DNA.


Assuntos
Antineoplásicos/química , DNA/química , Elipticinas/química , Substâncias Intercalantes/química , Pareamento de Bases , Cristalografia por Raios X/métodos
13.
Res Microbiol ; 153(4): 199-204, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12066890

RESUMO

Type IV secretory systems are transmembrane bacterial multiprotein complexes. They are pivotal for conjugation, bacterial-induced plant tumour formation, toxin secretion and mammalian pathogen intracellular activity. These systems are involved in the spread of antibiotic resistance genes among bacteria by enabling conjugative DNA transfer. When such translocons transport DNA, they require the assistance of multimeric integral inner membrane proteins, the type IV coupling proteins. Its structural prototype is plasmid R388 TrwB protein, responsible for coupling the relaxosome with the DNA transport apparatus during bacterial conjugation. Its monomeric molecular structure is reminiscent of ring helicases and AAA ATPases. The quaternary structure is made up by six equivalent protomers featuring a flattened sphere resembling F1-ATPase, with a central channel traversing the particle, thus connecting cytoplasm and periplasm.


Assuntos
Bactérias/genética , Proteínas de Bactérias/química , Conjugação Genética , Proteínas de Ligação a DNA/química , Proteínas de Escherichia coli , Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Moleculares , Plasmídeos
14.
J Biol Chem ; 277(30): 27273-81, 2002 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-12016211

RESUMO

Zyme/protease M/neurosin/human kallikrein 6 (hK6) is a member of the human kallikrein family of trypsin-like serine proteinases and was originally identified as being down-regulated in metastatic breast and ovarian tumors when compared with corresponding primary tumors. Recent evidence suggests that hK6 may serve as a circulating tumor marker in ovarian cancers. In addition, it was described in the brain of Parkinson's disease and Alzheimer's disease patients, where it is implicated in amyloid precursor protein processing. It is thus a biomarker for these diseases. To examine the mechanism of activation of hK6, we have solved the structure of its proform, the first of a human kallikrein family member. The proenzyme displays a fold that exhibits chimeric features between those of trypsinogen and other family members. It lacks the characteristic "kallikrein loop" and forms the six disulfide bridges of trypsin. Pro-hK6 displays a completely closed specificity pocket and a unique conformation of the regions involved in structural rearrangements upon proteolytic cleavage activation. This points to a novel activation mechanism, which could be extrapolated to other human kallikreins.


Assuntos
Calicreínas/química , Calicreínas/genética , Sequência de Aminoácidos , Sítios de Ligação , Encéfalo/metabolismo , Cristalografia por Raios X , Dissulfetos , Elétrons , Éxons , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
15.
Nat Struct Biol ; 9(1): 57-60, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11731803

RESUMO

Cryptolepine, a naturally occurring indoloquinoline alkaloid used as an antimalarial drug in Central and Western Africa, has been found to bind to DNA in a formerly unknown intercalation mode. Evidence from competition dialysis assays demonstrates that cryptolepine is able to bind CG-rich sequences containing nonalternating CC sites. Here we show that cryptolepine interacts with the CC sites of the DNA fragment d(CCTAGG)(2) in a base-stacking intercalation mode. This is the first DNA intercalator complex, from approximately 90 solved by X-ray crystallography, to bind a nonalternating (pyrimidine-pyrimidine) DNA sequence. The asymmetry of the drug induces a perfect stacking with the asymmetric site, allowing for the stability of the complex in the absence of hydrogen bonding interactions. The crystal structure of this antimalarial drug-DNA complex provides evidence for the first nonalternating intercalation and, as such, provides a basis for the design of new anticancer or antimalarial drugs.


Assuntos
Alcaloides/química , Alcaloides/metabolismo , Antimaláricos/metabolismo , Citosina/metabolismo , DNA/química , DNA/metabolismo , Indóis/química , Indóis/metabolismo , Substâncias Intercalantes/metabolismo , Quinolinas/química , Quinolinas/metabolismo , Antimaláricos/química , Sequência de Bases , Sítios de Ligação , Cristalografia por Raios X , DNA/genética , Diálise , Alcaloides Indólicos , Substâncias Intercalantes/química , Modelos Moleculares , Conformação Molecular , Conformação de Ácido Nucleico , Plantas Medicinais/química , Soluções
16.
EMBO Rep ; 3(1): 88-94, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11751578

RESUMO

The macrophage infectivity potentiator protein from Trypanosoma cruzi (TcMIP) is a major virulence factor secreted by the etiological agent of Chagas' disease. It is functionally involved in host cell invasion. We have determined the three-dimensional crystal structure of TcMIP at 1.7 A resolution. The monomeric protein displays a peptidyl-prolyl cis-trans isomerase (PPIase) core, encompassing the characteristic rotamase hydrophobic active site, thus explaining the strong inhibition of TcMIP by the immunosuppressant FK506 and related drugs. In TcMIP, the twisted beta-sheet of the core is extended by an extra beta-strand, preceded by a long, exposed N-terminal alpha-helix, which might be a target recognition element. An invasion assay shows that the MIP protein from Legionella pneumophila (LpMIP), which has an equivalent N-terminal alpha-helix, can substitute for TcMIP. An additional exposed alpha-helix, this one unique to TcMIP, is located in the C-terminus of the protein. The high-resolution structure reported here opens the possibility for the design of new inhibitory drugs that might be useful for the clinical treatment of American trypanosomiasis.


Assuntos
Imunofilinas/química , Proteínas de Membrana/química , Peptidilprolil Isomerase/química , Proteínas de Protozoários/química , Trypanosoma cruzi/química , Sequência de Aminoácidos , Animais , Proteínas de Bactérias , Sítios de Ligação , Humanos , Macrófagos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Proteínas de Protozoários/fisiologia , Homologia de Sequência , Homologia de Sequência de Aminoácidos , Proteínas de Ligação a Tacrolimo/química , Trypanosoma cruzi/enzimologia , Trypanosoma cruzi/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA