Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem ; 57: 116613, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35092894

RESUMO

In a screening using our unique natural product library, the C-nucleoside antibiotic formycin A, which exerts strong anti-influenza virus activity, was rediscovered. Aiming to develop a new type of anti-influenza virus drug, we synthesized new derivatives of formycin and evaluated its anti-influenza virus activity. Structural modifications were focused on the base moiety and sugar portion, respectively, and >40 novel formycin derivatives were synthesized. Modification of the C-7 position of the pyrazolopyrimidine ring strongly contributed to improve the activity. In particular, excellent anti-influenza virus activity was observed in the NHMe (10), SMe (12), and SeMe (15) derivatives, in which heteroatoms were introduced. In addition, in the modification of the sugar moiety, the presence of a hydroxyl group and its stereochemistry greatly affected both the expression and intensity of the activity. Furthermore, the evaluation results of the 7-SEt derivative (29) and the 2'-modified derivative (59) suggested that structural modifications may reduce cytotoxicity.


Assuntos
Antivirais/farmacologia , Formicinas/farmacologia , Orthomyxoviridae/efeitos dos fármacos , Antivirais/síntese química , Antivirais/química , Relação Dose-Resposta a Droga , Formicinas/síntese química , Formicinas/química , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade
2.
Chem Commun (Camb) ; 55(96): 14502-14505, 2019 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-31730149

RESUMO

ForI is a PLP-dependent enzyme from the biosynthetic pathway of the C-nucleoside antibiotic formycin. Cycloserine is thought to inhibit PLP-dependent enzymes by irreversibly forming a PMP-isoxazole. We now report that ForI forms novel PMP-diketopiperazine derivatives following incubation with both d and l cycloserine. This unexpected result suggests chemical diversity in the chemistry of cycloserine inhibition.


Assuntos
Proteínas de Bactérias/metabolismo , Dicetopiperazinas/química , Formicinas/biossíntese , Fosfato de Piridoxal/química , Piridoxamina/análogos & derivados , Transaminases/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Biocatálise , Domínio Catalítico , Ciclosserina/química , Dicetopiperazinas/metabolismo , Formicinas/química , Concentração de Íons de Hidrogênio , Piridoxamina/química , Piridoxamina/metabolismo , Streptomyces/química , Streptomyces/metabolismo , Transaminases/antagonistas & inibidores , Transaminases/genética
3.
J Am Chem Soc ; 141(15): 6127-6131, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30942582

RESUMO

Formycin A is a potent purine nucleoside antibiotic with a C-glycosidic linkage between the ribosyl moiety and the pyrazolopyrimidine base. Herein, a cosmid is identified from the Streptomyces kaniharaensis genome library that contains the for gene cluster responsible for the biosynthesis of formycin. Subsequent gene deletion experiments and in vitro characterization of the forBCH gene products established their catalytic functions in formycin biosynthesis. Results also demonstrated that PurH from de novo purine biosynthesis plays a key role in pyrazolopyrimidine formation during biosynthesis of formycin A. The participation of PurH in both pathways represents a good example of how primary and secondary metabolism are interlinked.


Assuntos
Formicinas/biossíntese , Purinas/biossíntese , Pirazóis/metabolismo , Pirimidinas/biossíntese , Streptomyces/química , Formicinas/química , Formicinas/metabolismo , Conformação Molecular , Família Multigênica , Purinas/química , Pirazóis/química , Pirimidinas/química , Estereoisomerismo , Streptomyces/genética , Streptomyces/metabolismo
4.
Chem Biodivers ; 16(4): e1900012, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30773842

RESUMO

Two lipophilic derivatives of formycin A (1) and formycin B (5) carrying an O-2',3'-(ethyl levulinate) ketal group have been prepared. These were base-alkylated at N(1) (for 1) and N(1) and N(6) (for 5) with both isopentenyl and all-trans-farnesyl residues. Upon the prenylation, side reactions were observed, resulting in the formation of nucleolipids with a novel tricyclic nucleobase (→4a, 4b). In the case of formycin B, O-2',3'-(ethyl levulinate) (6) farnesylation gave the double prenylated nucleolipid 7. All new compounds were characterized by 1 H-, 13 C-, UV/VIS and fluorescence spectroscopy, by ESI-MS spectrometry and/or by elemental analysis. Log P determinations between water and octanol as well as water and cyclohexane of a selection of compounds allowed qualitative conclusions concerning their potential blood-brain barrier passage efficiency. All compounds were investigated in vitro with respect to their cytotoxic activity toward rat malignant neuroectodermal BT4Ca as well as against a series of human glioblastoma cell lines (GOS 3, U-87 MG and GBM 2014/42). In order to differentiate between anticancer and side effects of the novel nucleolipids, we also studied their activity on PMA-differentiated human THP-1 macrophages. Here, we show that particularly the formycin A derivative 3b possesses promising antitumor properties in several cancer cell lines with profound cytotoxic effects partly on human glioblastoma cells, with a higher efficacy than the chemotherapeutic drug 5-fluorouridine.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Formicinas/farmacologia , Animais , Antibacterianos/síntese química , Antibacterianos/química , Antineoplásicos Fitogênicos/síntese química , Antineoplásicos Fitogênicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Formicinas/síntese química , Formicinas/química , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Ratos , Relação Estrutura-Atividade , Células Tumorais Cultivadas
5.
Biophys Chem ; 230: 99-108, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28947300

RESUMO

The aim of this study is threefold: (1) augmentation of the knowledge of the E. coli PNP binding mechanism; (2) explanation of the previously observed 'lack of FRET' phenomenon and (3) an introduction of the correction (modified method) for FRET efficiency calculation in the PNP-FA complexes. We present fluorescence studies of the two E. coli PNP mutants (F159Y and F159A) with formycin A (FA), that indicate that the aromatic amino acid is indispensable in the nucleotide binding, additional hydroxyl group at position 159 probably enhances the strength of binding and that the amino acids pair 159-160 has a great impact on the spectroscopic properties of the enzyme. The experiments were carried out in hepes and phosphate buffers, at pH7 and 8.3. Two methods, a conventional and a modified one, that utilizes the dissociation constant, for calculations of the energy transfer efficiency (E) and the acceptor-to-donor distance (r) between FA and the Tyr (energy donor) were employed. Total difference spectra were calculated for emission spectra (λex 280nm, 295nm, 305nm and 313nm) for all studied systems. Time-resolved techniques allowed to conclude the existence of a specific structure formed by amino acids at positions 159 and 160. The results showed an unexpected pattern change of FRET in the mutants, when compared to the wild type enzyme and a probable presence of a structure created between 159 and 160 residue, that might influence the binding efficiency. Additionally, we confirmed the indispensable role of the modification of the FRET efficiency (E) calculation on the fraction of enzyme saturation in PNP-FA systems.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Formicinas/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Transferência Ressonante de Energia de Fluorescência , Formicinas/química , Concentração de Íons de Hidrogênio , Cinética , Mutagênese Sítio-Dirigida , Ligação Proteica , Estrutura Terciária de Proteína , Purina-Núcleosídeo Fosforilase/química , Purina-Núcleosídeo Fosforilase/genética , Espectrometria de Fluorescência , Temperatura
6.
Proc Natl Acad Sci U S A ; 113(48): 13756-13761, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27856757

RESUMO

MTAN (5'-methylthioadenosine nucleosidase) catalyzes the hydrolysis of the N-ribosidic bond of a variety of adenosine-containing metabolites. The Helicobacter pylori MTAN (HpMTAN) hydrolyzes 6-amino-6-deoxyfutalosine in the second step of the alternative menaquinone biosynthetic pathway. Substrate binding of the adenine moiety is mediated almost exclusively by hydrogen bonds, and the proposed catalytic mechanism requires multiple proton-transfer events. Of particular interest is the protonation state of residue D198, which possesses a pKa above 8 and functions as a general acid to initiate the enzymatic reaction. In this study we present three corefined neutron/X-ray crystal structures of wild-type HpMTAN cocrystallized with S-adenosylhomocysteine (SAH), Formycin A (FMA), and (3R,4S)-4-(4-Chlorophenylthiomethyl)-1-[(9-deaza-adenin-9-yl)methyl]-3-hydroxypyrrolidine (p-ClPh-Thio-DADMe-ImmA) as well as one neutron/X-ray crystal structure of an inactive variant (HpMTAN-D198N) cocrystallized with SAH. These results support a mechanism of D198 pKa elevation through the unexpected sharing of a proton with atom N7 of the adenine moiety possessing unconventional hydrogen-bond geometry. Additionally, the neutron structures also highlight active site features that promote the stabilization of the transition state and slight variations in these interactions that result in 100-fold difference in binding affinities between the DADMe-ImmA and ImmA analogs.


Assuntos
Formicinas/química , Helicobacter pylori/enzimologia , Purina-Núcleosídeo Fosforilase/química , S-Adenosil-Homocisteína/química , Adenina/análogos & derivados , Adenina/química , Domínio Catalítico/genética , Cristalografia por Raios X , Desoxiadenosinas/química , Helicobacter pylori/química , Ligação de Hidrogênio , Modelos Moleculares , Nêutrons , Ligação Proteica , Prótons , Purina-Núcleosídeo Fosforilase/genética , Pirrolidinas/química , Especificidade por Substrato , Tionucleosídeos/química
7.
Bioorg Med Chem Lett ; 26(19): 4720-4723, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27575480

RESUMO

Strontium fructose 1,6-diphosphate (FDP-Sr) is a new strontium-containing compound. The primary aim of this study was to clarify whether the structure component of FDP-Sr, FDP could benefit the protective effect of Sr (II) against oxidative stress induced apoptosis, and meanwhile to further explore the important role of Wnt/ß-catenin signaling in the anti-apoptosis effect of FDP-Sr in response to oxidative stress induced by H2O2 in an osteoblastic MC3T3-E1 cell line. Results showed that FDP-Sr could improve the osteoblastic differentiation under oxidative stress with induced cell proliferation and improved mineralization. The inhibition effect of FDP-Sr on cell apoptosis induced by H2O2 was proved by reduced reactive oxygen species production and activated caspase3. Under oxidative stress, mRNA and protein levels of phospho-ß-catenin reduced, while ß-catenin increased in the FDP-Sr treatment cell, leaded to the up-regulations of Runx2 and OPG at both mRNA and protein levels, finally improved the differentiation of osteoblasts. By the engagement of Wnt/ß-catenin pathway's inhibitor (XAV-939), the protective effects of FDP-Sr on osteoblastic differentiation against oxidative stress were repressed along with inhibited wnt/ß-catenin signaling and reduced mRNA and protein levels of Runx2 and OPG. In conclusion, FDP-Sr was demonstrated to protect osteoblast differentiation from oxidative damage induced by H2O2 through up-regulation of Wnt/ß-catenin signaling, and FDP in FDP-Sr was able to directly improve the oxidative stress injury through its ROS scavenging ability.


Assuntos
Formicinas/química , Estresse Oxidativo/efeitos dos fármacos , Ribonucleotídeos/química , Transdução de Sinais , Proteínas Wnt/metabolismo , Células 3T3 , Animais , Camundongos
8.
J Mol Model ; 21(4): 75, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25754135

RESUMO

Predicting FRET pathways in proteins using computer simulation techniques is very important for reliable interpretation of experimental data. A novel and relatively simple methodology has been developed and applied to purine nucleoside phosphorylase (PNP) complexed with a fluorescent ligand - formycin A (FA). FRET occurs between an excited Tyr residue (D*) and FA (A). This study aims to interpret experimental data that, among others, suggests the absence of FRET for the PNPF159A mutant in complex with FA, based on novel theoretical methodology. MD simulations for the protein molecule containing D*, and complexed with A, are carried out. Interactions of D* with its molecular environment are accounted by including changes of the ESP charges in S1, compared to S0, and computed at the SCF-CI level. FRET probability W F depends on the inverse six-power of the D*-A distance, R da . The orientational factor 0 < k(2) < 4 between D* and A is computed and included in the analysis. Finally W F is time-averaged over the MD trajectories resulting in its mean value. The red-shift of the tyrosinate anion emission and thus lack of spectral overlap integral and thermal energy dissipation are the reasons for the FRET absence in the studied mutants at pH 7 and above. The presence of the tyrosinate anion results in a competitive energy dissipation channel and red-shifted emission, thus in consequence in the absence of FRET. These studies also indicate an important role of the phenyl ring of Phe159 for FRET in the wild-type PNP, which does not exist in the Ala159 mutant, and for the effective association of PNP with FA. In a more general context, our observations point out very interesting and biologically important properties of the tyrosine residue in its excited state, which may undergo spontaneous deprotonation in the biomolecular systems, resulting further in unexpected physical and/or biological phenomena. Until now, this observation has not been widely discussed in the literature.


Assuntos
Escherichia coli/enzimologia , Formicinas/química , Purina-Núcleosídeo Fosforilase/química , Purina-Núcleosídeo Fosforilase/genética , Sítios de Ligação , Simulação por Computador , Escherichia coli/genética , Transferência Ressonante de Energia de Fluorescência , Formicinas/farmacologia , Ligantes , Mutação , Conformação Proteica , Purina-Núcleosídeo Fosforilase/metabolismo , Espectrometria de Fluorescência , Especificidade por Substrato
9.
FEBS Lett ; 586(7): 967-71, 2012 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-22569248

RESUMO

Purine nucleoside phosphorylase (PNP) from Escherichia coli is a homohexamer that catalyses the phosphorolytic cleavage of the glycosidic bond of purine nucleosides. The first crystal structure of the ternary complex of this enzyme (with a phosphate ion and formycin A), which is biased by neither the presence of an inhibitor nor sulfate as a precipitant, is presented. The structure reveals, in some active sites, an unexpected and never before observed binding site for phosphate and exhibits a stoichiometry of two phosphate molecules per enzyme subunit. Moreover, in these active sites, the phosphate and nucleoside molecules are found not to be in direct contact. Rather, they are bridged by three water molecules that occupy the "standard" phosphate binding site.


Assuntos
Antineoplásicos/metabolismo , Inibidores Enzimáticos/metabolismo , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Formicinas/metabolismo , Fosfatos/metabolismo , Purina-Núcleosídeo Fosforilase/química , Antineoplásicos/química , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/química , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Formicinas/química , Cinética , Ligantes , Modelos Moleculares , Concentração Osmolar , Fosfatos/química , Conformação Proteica , Purina-Núcleosídeo Fosforilase/antagonistas & inibidores , Purina-Núcleosídeo Fosforilase/genética , Purina-Núcleosídeo Fosforilase/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrometria de Fluorescência , Titulometria , Água/química , Água/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-18066912

RESUMO

Phosphorolysis of 7-methylguanosine by calf spleen purine nucleoside phosphorylase (PNP) is weakly inhibited, uncompetitively, by Formycin B (FB) with Ki = 100 micro M and more effectively by its aglycone (7KPP), IC50 35-100 micro M. In striking contrast, 7KPP inhibits the reverse reaction (synthesis of 8-azaguanosine from 8-azaguanine) competitively, with Ki approximately 2-4 micro M. Formycin B forms only a weakly fluorescent complex with PNP, and 7KPP even less so, indicating that both ligands bind as the neutral, not anionic, forms. 7KPP is a rare example of a PNP non-substrate inhibitor of both the phosphorolytic and reverse synthetic pathways.


Assuntos
Formicinas/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Baço/enzimologia , Animais , Bovinos , Formicinas/química , Guanosina/análogos & derivados , Guanosina/química , Guanosina/metabolismo , Cinética , Pirimidinas/química , Pirimidinas/metabolismo , Espectrometria de Fluorescência , Especificidade por Substrato
11.
Biophys Chem ; 123(2-3): 146-53, 2006 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-16765509

RESUMO

Fluorescence decays in protein-ligand complexes are described by a new efficient model of continuous distribution of fluorescence lifetimes, and compared with multi-exponential models. Resulted analytical power-like decay function provides good fits to highly complex fluorescence kinetics. Moreover, this is a manifestation of so-called Tsallis q-exponential function, which is suitable for description of the systems with long-range interactions, memory effect, as well as with fluctuations of the characteristic lifetime of fluorescence. The proposed decay function was used to study effect of the interaction of E. coli purine nucleoside phosphorylase (PNP-I, the product of the deoD gene) with its specific inhibitor, viz. formycin A (FA), on fluorescence decays of ligand and enzyme tyrosine residues, in the presence of orthophosphate (P(i), a natural co-substrate). The power-like function provides new information about enzyme-ligand complex formation based on the excited state mean lifetime, heterogeneity parameter (q) and a number (N) of decay channels obtained from the variance of gamma distribution of fluorescence decay rates. With FA, which exists as a 85:15 mixture of the N(1)-H and N(2)-H tautomeric forms in aqueous solution, fluorescence intensity decay (lambda(exc)/lambda(em) 270/335 nm) is described by q approximately 1 and N approximately 200. Consequently power-like decay function converges to the single-exponential form, and lifetime distribution to the Dirac delta function. In contrast, selective excitation of the N(2)-H tautomer at higher wavelength led to a highly heterogenic fluorescence decay characterized by q>1 and 10-fold lower number of decay channels. Heterogeneity of fluorescence decays of both PNP-I and FA is enhanced by PNP-FA-P(i) complex formation, reflecting a shift of the tautomeric equilibrium of FA in favor of the N(2)-H species, and fluorescence resonance energy transfer (FRET) from protein tyrosine residue (Tyr160) to the bound N(2)-H tautomer. Moreover, proposed model is simple, and objectively describes heterogeneous nature of studied systems.


Assuntos
Proteínas de Bactérias/química , Transferência Ressonante de Energia de Fluorescência/métodos , Corantes Fluorescentes/química , Formicinas/química , Interpretação de Imagem Assistida por Computador/métodos , Purina-Núcleosídeo Fosforilase/química , Escherichia coli/enzimologia , Fluorescência , Ligantes , Modelos Teóricos , Estrutura Molecular , Sensibilidade e Especificidade , Relação Estrutura-Atividade , Tirosina/química
12.
Artigo em Inglês | MEDLINE | ID: mdl-16248001

RESUMO

Chemical synthesis of a series of novel dinucleoside cap analogues, m7GpppN, where N is formycin A, 3'-O-methylguanosine, 9-beta-D-arabinofuranosyladenine, and isoguanosine, has been performed using our new methodology. The key reactions of pyrophosphate bonds formation were achieved in anhydrous dimethylformamide solutions employing the catalytic properties of zinc salts. Structures of the new cap analogues were confirmed by 1H NMR and 31p NMR spectra. The binding affinity of the new cap analogues for murine eIF4E(28-217) were determined spectroscopically showing the highest association constant for the analogue that contains formycin A.


Assuntos
Fator de Iniciação 4E em Eucariotos/química , Fator de Iniciação 4E em Eucariotos/genética , Nucleosídeos/química , Análogos de Capuz de RNA , Adenosina , Animais , Relação Dose-Resposta a Droga , Formicinas/química , Guanosina/química , Cinética , Espectroscopia de Ressonância Magnética , Camundongos , Modelos Químicos , Nucleotídeos/química , Ligação Proteica , Espectrofotometria , Termodinâmica , Vidarabina/química
13.
J Biol Chem ; 280(23): 22318-25, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15817485

RESUMO

Trichomonas vaginalis is an anaerobic protozoan parasite that causes trichomoniasis, a common sexually transmitted disease with worldwide impact. One of the pivotal enzymes in its purine salvage pathway, purine nucleoside phosphorylase (PNP), shows physical properties and substrate specificities similar to those of the high molecular mass bacterial PNPs but differing from those of human PNP. While carrying out studies to identify inhibitors of T. vaginalis PNP (TvPNP), we discovered that the nontoxic nucleoside analogue 2-fluoro-2'-deoxyadenosine (F-dAdo) is a "subversive substrate." Phosphorolysis by TvPNP of F-dAdo, which is not a substrate for human PNP, releases highly cytotoxic 2-fluoroadenine (F-Ade). In vitro studies showed that both F-dAdo and F-Ade exert strong inhibition of T. vaginalis growth with estimated IC(50) values of 106 and 84 nm, respectively, suggesting that F-dAdo might be useful as a potential chemotherapeutic agent against T. vaginalis. To understand the basis of TvPNP specificity, the structures of TvPNP complexed with F-dAdo, 2-fluoroadenosine, formycin A, adenosine, inosine, or 2'-deoxyinosine were determined by x-ray crystallography with resolutions ranging from 2.4 to 2.9 A. These studies showed that the quaternary structure, monomer fold, and active site are similar to those of Escherichia coli PNP. The principal active site difference is at Thr-156, which is alanine in E. coli PNP. In the complex of TvPNP with F-dAdo, Thr-156 causes the purine base to tilt and shift by 0.5 A as compared with the binding scheme of F-dAdo in E. coli PNP. The structures of the TvPNP complexes suggest opportunities for further improved subversive substrates beyond F-dAdo.


Assuntos
Adenina/análogos & derivados , Adenosina/análogos & derivados , Inosina/análogos & derivados , Purina-Núcleosídeo Fosforilase/química , Trichomonas vaginalis/metabolismo , Adenina/química , Adenosina/química , Animais , Sítios de Ligação , Catálise , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Elétrons , Formicinas/química , Humanos , Hidrólise , Concentração Inibidora 50 , Inosina/química , Ligantes , Modelos Químicos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/química , Especificidade por Substrato , Treonina/química , Fatores de Tempo , Raios X
14.
Structure ; 12(8): 1383-94, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15296732

RESUMO

AMP nucleosidase (AMN) catalyzes the hydrolysis of AMP to form adenine and ribose 5-phosphate. The enzyme is found only in prokaryotes, where it plays a role in purine nucleoside salvage and intracellular AMP level regulation. Enzyme activity is stimulated by ATP and suppressed by phosphate. The structure of unliganded AMN was determined at 2.7 A resolution, and structures of the complexes with either formycin 5'-monophosphate or inorganic phosphate were determined at 2.6 A and 3.0 A resolution, respectively. AMN is a biological homohexamer, and each monomer is composed of two domains: a catalytic domain and a putative regulatory domain. The overall topology of the catalytic domain and some features of the substrate binding site resemble those of the nucleoside phosphorylases, demonstrating that AMN is a new member of the family. The structure of the regulatory domain consists of a long helix and a four-stranded sheet and has a novel topology.


Assuntos
Formicinas/química , Modelos Moleculares , N-Glicosil Hidrolases/química , Pentosiltransferases/química , Ribonucleotídeos/química , Ribosemonofosfatos/química , Sequência de Aminoácidos , Domínio Catalítico , Cristalografia por Raios X , Escherichia coli/enzimologia , Dados de Sequência Molecular , Fosfatos/química , Homologia de Sequência de Aminoácidos
15.
Biochemistry ; 43(18): 5159-69, 2004 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-15122881

RESUMO

The development of new and effective antiprotozoal drugs has been a difficult challenge because of the close similarity of the metabolic pathways between microbial and mammalian systems. 5'-Methylthioadenosine/S-adenosylhomocysteine (MTA/AdoHcy) nucleosidase is thought to be an ideal target for therapeutic drug design as the enzyme is present in many microbes but not in mammals. MTA/AdoHcy nucleosidase (MTAN) irreversibly depurinates MTA or AdoHcy to form adenine and the corresponding thioribose. The inhibition of MTAN leads to a buildup of toxic byproducts that affect various microbial pathways such as quorum sensing, biological methylation, polyamine biosynthesis, and methionine recycling. The design of nucleosidase-specific inhibitors is complicated by its structural similarity to the human MTA phosphorylase (MTAP). The crystal structures of human MTAP complexed with formycin A and 5'-methylthiotubercidin have been solved to 2.0 and 2.1 A resolution, respectively. Comparisons of the MTAP and MTAN inhibitor complexes reveal size and electrostatic potential differences in the purine, ribose, and 5'-alkylthio binding sites, which account for the substrate specificity and reactions catalyzed. In addition, the differences between the two enzymes have allowed the identification of exploitable regions that can be targeted for the development of high-affinity nucleosidase-specific inhibitors. Sequence alignments of Escherichia coli MTAN, human MTAP, and plant MTA nucleosidases also reveal potential structural changes to the 5'-alkylthio binding site that account for the substrate preference of plant MTA nucleosidases.


Assuntos
Inibidores Enzimáticos/síntese química , N-Glicosil Hidrolases/antagonistas & inibidores , N-Glicosil Hidrolases/química , Purina-Núcleosídeo Fosforilase/antagonistas & inibidores , Purina-Núcleosídeo Fosforilase/química , Tubercidina/análogos & derivados , Nucleotídeos de Adenina/química , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Formicinas/química , Humanos , Dados de Sequência Molecular , Fosfatos/química , Nucleotídeos de Purina/química , Ribose/química , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Sulfatos/química , Tionucleosídeos/química , Tubercidina/química
16.
Artigo em Inglês | MEDLINE | ID: mdl-15043159

RESUMO

The Mitsunobu reaction was applied to prepare, in one step, purine N(3),5'-cyclonucleosides 10a-d. A subsequent ring opening in the ribose moiety of the resultant N(3),5'-nucleosides by sodium periodate led to the corresponding N(3),5'-cyclo-2',3'-seconucleosides. These products consist of 5-, 6-, and 7-membered tricyclic system which is the basic skeleton of TIBO derivatives, known antiviral agents.


Assuntos
Nucleosídeos de Purina/síntese química , Benzodiazepinas/síntese química , Benzodiazepinas/química , Formicinas/química , Imidazóis/síntese química , Imidazóis/química
17.
Eur Biophys J ; 33(5): 377-85, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-14655027

RESUMO

Fluorescence and phosphorescence emission spectroscopy were employed to study the interaction of Escherichia coli purine nucleoside phosphorylase (PNP) with its specific inhibitor, formycin A (FA), a close structural analogue of adenosine (natural substrate), in the absence and presence of phosphate (P(i), substrate). Formation of enzyme-FA complexes led to marked quenching of enzyme tyrosine intrinsic fluorescence and phosphorescence, with concomitant increases in fluorescence and phosphorescence of FA. Fluorescence resonance energy transfer from the protein Tyr160 residue to the FA base moiety was identified as a major mechanism of protein fluorescence quenching, increased by addition of P(i). The effects of enzyme-FA interactions on the nucleoside excitation and emission spectra for fluorescence and phosphorescence revealed shifts in the tautomeric equilibrium of the bound FA, i.e. from the N(1)-H tautomer (predominant in solution) to the N(2)-H form, enhanced by the presence of P(i). The latter was confirmed by enzyme-ligand dissociation constant ( K(d)) values of 5.9+/-0.4 and 2.1+/-0.3 microM in the absence and presence of P(i), respectively. Addition of glycerol (80%, v/v) led to a lower enzyme affinity ( K(d) approximately 70 microM), without changes in binding stoichiometry. Enzyme-FA complex formation led to a higher increase of the fluorescence than the phosphorescence band of the ligand, consistent with the fact that the N(2)-H tautomer is characterized by a weaker phosphorescence than the N(1)-H tautomeric form. These results show, for the first time, the application of phosphorescence spectroscopy to the identification of the tautomeric form of the inhibitor bound by the enzyme.


Assuntos
Algoritmos , Escherichia coli/enzimologia , Formicinas/química , Purina-Núcleosídeo Fosforilase/química , Espectrometria de Fluorescência/métodos , Sítios de Ligação , Ativação Enzimática , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/química , Transferência Ressonante de Energia de Fluorescência/métodos , Formicinas/análise , Formicinas/classificação , Isomerismo , Cinética , Substâncias Macromoleculares/análise , Substâncias Macromoleculares/química , Ligação Proteica , Purina-Núcleosídeo Fosforilase/análise , Purina-Núcleosídeo Fosforilase/antagonistas & inibidores
18.
Biophys J ; 85(1): 589-98, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12829513

RESUMO

A power-like decay function, characterized by the mean excited-state lifetime and relative variance of lifetime fluctuation around the mean value, was applied in analysis of fluorescence decays measured with the aid of time-correlated single photon counting. We have examined the fluorescence decay, in neutral aqueous medium, of tyrosine (L-tyrosine and N-acetyl-L-tyrosinamide), and of the tyrosine residues in a tryptophan-free protein, the enzyme purine nucleoside phosphorylase from Escherichia coli in a complex with formycin A (an inhibitor), and orthophosphate (a co-substrate). Tryptophan fluorescence decay was examined in neutral aqueous medium for L-tryptophan, N-acetyl-L-tryptophanamide, and for two tryptophan residues in horse liver alcohol dehydrogenase. To detect solvent effect, fluorescence decay of Nz-acetyl-L-tryptophanamide in aqueous medium was compared with that in dioxan. Hitherto, complex fluorescence decays have usually been analyzed with the aid of a multiexponential model, but interpretation of the individual exponential terms (i.e., pre-exponential amplitudes and fluorescence lifetimes), has not been adequately characterized. In such cases the intensity decays were also analyzed in terms of the lifetime distribution as a consequence of an interaction of fluorophore with environment. We show that the power-like decay function, which can be directly obtained from the gamma distribution of fluorescence lifetimes, is simpler and provides good fits to highly complex fluorescence decays as well as to a purely single-exponential decay. Possible interpretation of the power-like model is discussed.


Assuntos
Fluorescência , Formicinas/química , Modelos Químicos , Espectrometria de Fluorescência/métodos , Triptofano/análogos & derivados , Triptofano/química , Tirosina/química , Simulação por Computador , Formicinas/análise , Formicinas/metabolismo , Meia-Vida , Luz , Triptofano/análise , Triptofano/efeitos da radiação , Tirosina/análise , Tirosina/efeitos da radiação
19.
J Mol Biol ; 315(3): 351-71, 2002 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-11786017

RESUMO

The crystal structure of the ternary complex of hexameric purine nucleoside phosphorylase (PNP) from Escherichia coli with formycin A derivatives and phosphate or sulphate ions is determined at 2.0 A resolution. The hexamer is found as a trimer of unsymmetric dimers, which are formed by pairs of monomers with active sites in different conformations. The conformational difference stems from a flexible helix (H8: 214-236), which is continuous in one conformer, and segmented in the other. With the continuous helix, the entry into the active site pocket is wide open, and the ligands are bound only loosely ("open" or "loose binding" conformation). By segmentation of the helix (H8: 214-219 and H8': 223-236, separated by a gamma-turn), the entry into the active site is partially closed, the pocket is narrowed and the ligands are bound much more tightly ("closed" or "tight binding" conformation). Furthermore, the side-chain of Arg217 is carried by the moving helix into the active site. This residue, conserved in all homologous PNPs, plays an important role in the proposed catalytic mechanism. In this mechanism, substrate binding takes place in the open, and and the catalytic action occurs in the closed conformation. Catalytic action involves protonation of the purine base at position N7 by the side-chain of Asp204, which is initially in the acid form. The proton transfer is triggered by the Arg217 side-chain which is moved by the conformation change into hydrogen bond distance to Asp204. The mechanism explains the broad specificity of E. coli PNP, which allows 6-amino as well as 6-oxo-nucleosides as substrates. The observation of two kinds of binding sites is fully in line with solution experiments which independently observe strong and weak binding sites for phosphate as well as for the nucleoside inhibitor.


Assuntos
Escherichia coli/enzimologia , Purina-Núcleosídeo Fosforilase/química , Purina-Núcleosídeo Fosforilase/metabolismo , Sítios de Ligação , Catálise , Cristalização , Cristalografia por Raios X , Dimerização , Fluorescência , Formicinas/química , Formicinas/metabolismo , Guanosina/química , Guanosina/metabolismo , Ligação de Hidrogênio , Inosina/química , Inosina/metabolismo , Cinética , Modelos Moleculares , Nucleosídeos/química , Nucleosídeos/metabolismo , Fosfatos/metabolismo , Estrutura Quaternária de Proteína , Purina-Núcleosídeo Fosforilase/antagonistas & inibidores , Relação Estrutura-Atividade , Sulfatos/metabolismo , Titulometria
20.
J Biomol Struct Dyn ; 19(2): 285-91, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11697733

RESUMO

Fluorescent tRNAs species with formycine in the 3'-terminal position (tRNA-CCF) were derived from Escherichia coli tRNA(Val). Thermus thermophilus tRNA(Aap) and Thermus thermophilus tRNA(Phe). The fluorescence of formycine was used to monitor the conformational changes at the 3'-terminus of tRNA caused by aminoacylation and hydrolysis of aminoacyl residue from aminoacyl-tRNAs. An increase of about 15% in the fluorescence intensity was observed after aminoacylation of the three tRNA-CCF. This change in fluorescence amplitude that is reversed by hydrolysis of the aminoacyl residue, does not depend on the structure of the amino acid or tRNA sequence. A local conformational change at the 3'-terminal formycine probably involving a partial destacking of the base moiety in the ACCF end takes place as a consequence of aminoacylation. A structural change at the 3'-terminus of tRNA induced by attachment and detachment of the acyl residue may be important in controlling the substrate/product relationship in reactions in which tRNA participates during protein biosynthesis.


Assuntos
Conformação de Ácido Nucleico , Aminoacil-RNA de Transferência/química , Cromatografia Líquida de Alta Pressão , Escherichia coli/química , Corantes Fluorescentes/química , Formicinas/química , Espectroscopia de Ressonância Magnética , RNA de Transferência de Ácido Aspártico/química , RNA de Transferência de Fenilalanina/química , RNA de Transferência de Valina/química , Ribonucleotídeos/química , Thermus thermophilus/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...