Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Molecules ; 27(3)2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35164019

RESUMO

Breast cancer is a major cause of death in women worldwide. In this study, 60 female rats were classified into 6 groups; negative control, α-aminophosphonates, arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one, DMBA, DMBA & α-aminophosphonates, and DMBA & arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one. New α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one were synthesized and elucidated by different spectroscopic and elemental analysis. Histopathological examination showed marked proliferation of cancer cells in the DMBA group. Treatment with α-aminophosphonates mainly decreased tumor mass. Bcl2 expression increased in DMBA-administered rats and then declined in the treated groups, mostly with α-aminophosphonates. The level of CA15-3 markedly declined in DMBA groups treated with α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one. Gene expression of GST-P, PCNA, PDK, and PIK3CA decreased in the DMBA group treated with α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one, whereas PIK3R1 and BAX increased in the DMBA group treated with α-aminophosphonates and arylidine derivatives of 3-acetyl-1-aminoquinolin-2(1H)-one. The molecular docking postulated that the investigated compounds can inhibt the Thymidylate synthase TM due to high hydrophobicity charachter.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Timidilato Sintase/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno , Animais , Antineoplásicos/farmacologia , Células CACO-2 , Simulação por Computador , Avaliação Pré-Clínica de Medicamentos , Feminino , Peixes , Humanos , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Modelos Moleculares , Simulação de Acoplamento Molecular , Terapia de Alvo Molecular/métodos , Organofosfonatos/síntese química , Organofosfonatos/química , Organofosfonatos/farmacologia , Organofosfonatos/uso terapêutico , Extratos Vegetais , Quinolinas/síntese química , Quinolinas/química , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Ratos , Timidilato Sintase/química
2.
Sci Rep ; 6: 27198, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27250901

RESUMO

Demonstrating a candidate drug's interaction with its target protein in live cells is of pivotal relevance to the successful outcome of the drug discovery process. Although thymidylate synthase (hTS) is an important anticancer target protein, the efficacy of the few anti-hTS drugs currently used in clinical practice is limited by the development of resistance. Hence, there is an intense search for new, unconventional anti-hTS drugs; there are approximately 1600 ongoing clinical trials involving hTS-targeting drugs, both alone and in combination protocols. We recently discovered new, unconventional peptidic inhibitors of hTS that are active against cancer cells and do not result in the overexpression of hTS, which is a known molecular source of resistance. Here, we propose an adaptation of the recently proposed tetracysteine-arsenic-binding-motif technology to detect and quantitatively characterize the engagement of hTS with one such peptidic inhibitor in cell lysates. This new model can be developed into a test for high-throughput screening studies of intracellular target-protein/small-molecule binding.


Assuntos
Arsênio/química , Cisteína/química , Inibidores Enzimáticos/química , Timidilato Sintase/química , Motivos de Aminoácidos , Arsênio/metabolismo , Domínio Catalítico , Cisteína/metabolismo , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Timidilato Sintase/antagonistas & inibidores
3.
Biosens Bioelectron ; 64: 36-42, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25189098

RESUMO

Thymidylate synthase (TS) and dihydrofolate reductase (DHFR) play essential role in DNA synthesis, repair and cell division by catalyzing two subsequent reactions in thymidylate biosynthesis cycle. The lack of either enzyme leads to thymineless death of the cell, therefore inhibition of the enzyme activity is a common and successful tool in cancer chemotherapy and treatment of other diseases. However, the detailed mechanism of thymidylate synthesis cycle, especially the interactions between cycle enzymes and its role remain unknown. In this paper we are the first to show that human TS and DHFR enzymes form a strong complex which might be essential for DNA synthesis. Using two unique biosensor techniques, both highly sensitive to biomolecular interactions, namely quartz crystal microbalance with dissipation monitoring (QCM-D) and microscale thermophoresis (MST) we have been able to determine DHFR-TS binding kinetic parameters such as the Kd value being below 10 µM (both methods), k(on) = 0.46 × 10(4) M(-1) s(-1) and k(off) = 0.024 s(-1) (QCM-D). We also calculated Gibbs free energy as in the order of -30 kJ/mol and DHFR/TS molar ratio pointing to binding of 6 DHFR monomers per 1 TS dimer (both methods). Moreover, our data from MST analysis have pointed to positive binding cooperativity in TS-DHFR complex formation. The results obtained with both methods are comparable and complementary.


Assuntos
Técnicas Biossensoriais/métodos , Complexos Multienzimáticos/metabolismo , Técnicas de Microbalança de Cristal de Quartzo/métodos , Tetra-Hidrofolato Desidrogenase/metabolismo , Timidilato Sintase/metabolismo , Humanos , Cinética , Complexos Multienzimáticos/química , Mapeamento de Interação de Proteínas/métodos , Tetra-Hidrofolato Desidrogenase/química , Termodinâmica , Timidilato Sintase/química
4.
J Mol Model ; 18(9): 4089-100, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22527276

RESUMO

Leishmaniasis, a multi-faceted ethereal disease is considered to be one of the World's major communicable diseases that demands exhaustive research and control measures. The substantial data on these protozoan parasites has not been utilized completely to develop potential therapeutic strategies against Leishmaniasis. Dihydrofolate reductase thymidylate synthase (DHFR-TS) plays a major role in the infective state of the parasite and hence the DHFR-TS based drugs remains of much interest to researchers working on Leishmaniasis. Although, crystal structures of DHFR-TS from different species including Plasmodium falciparum and Trypanosoma cruzi are available, the experimentally determined structure of the Leishmania major DHFR-TS has not yet been reported in the Protein Data Bank. A high quality three dimensional structure of L.major DHFR-TS has been modeled through the homology modeling approach. Carefully refined and the energy minimized structure of the modeled protein was validated using a number of structure validation programs to confirm its structure quality. The modeled protein structure was used in the process of structure based virtual screening to figure out a potential lead structure against DHFR TS. The lead molecule identified has a binding affinity of 0.51 nM and clearly follows drug like properties.


Assuntos
Antiprotozoários/análise , Antiprotozoários/farmacologia , Leishmania major/enzimologia , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/química , Biblioteca de Peptídeos , Tetra-Hidrofolato Desidrogenase/química , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/química , Interface Usuário-Computador , Aminoácidos/química , Domínio Catalítico , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Ligação de Hidrogênio/efeitos dos fármacos , Leishmania major/efeitos dos fármacos , Ligantes , Espectroscopia de Ressonância Magnética , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Termodinâmica
5.
Bioorg Med Chem Lett ; 18(12): 3628-31, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18513963

RESUMO

Three synthesized series of compounds based on a thiazolidine core allowed identification of potent inhibitors of thymidylate synthase X. The evaluation of the catalytic activity of the enzyme in the presence of these molecules revealed two distinct classes of compounds that inhibit ThyX with submicromolar concentrations, which could lead, after optimization, to effective inhibitors with potential biomedical interest.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Tiazolidinas/síntese química , Tiazolidinas/farmacologia , Timidilato Sintase/antagonistas & inibidores , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/química , Conformação Molecular , Estereoisomerismo , Relação Estrutura-Atividade , Tiazolidinas/química , Timidilato Sintase/química , Fatores de Tempo
6.
Curr Drug Targets ; 3(4): 297-309, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12102601

RESUMO

Thymidylate synthase (TS) is an essential enzyme in de novo synthesis of thymidylate, and is required for DNA synthesis and cell proliferation in the absence of exogenous thymidine. As a consequence, TS is a target for anticancer chemotherapy by several drugs, including 5-fluorouracil (5-FU) and raltitrexed (Tomudex), in treatment of colorectal and other tumors. TS overexpression due to increased gene transcription and mRNA translation can mediate drug resistance. Decreased cellular uptake and polyglutamylation of TS-targeting drugs (raltitrexed, for example), increased drug efflux, altered metabolism of cytotoxic drugs (for example, 5-FU), and other events can decrease the effectiveness of TS-targeting drugs. Recent preclinical and clinical studies have addressed the resistance problem by using combinations of different drugs that target TS, or by combining TS-targeting and non-TS-targeting drugs. Our approach has been to circumvent and/or prevent TS overexpression-mediated drug resistance by employing antisense oligodeoxynucleotides (ODNs) to downregulate TS mRNA and protein levels. These studies have revealed that targeting the 3' end of human TS mRNA downregulates TS mRNA and protein, inhibits cell proliferation, and sensitizes HeLa cells to raltitrexed, 5-FU, and 5-fluorodeoxyuridine (5-FUdR) in vitro (Ferguson et al., Br. J Pharmacol. 127, 1777-1786, 1999). In addition, growth of human HT29 colon carcinoma cell explants in immunocompromised mice is inhibited by antisense downregulation of TS (Berg et al., J. Pharmacol. Exp. Therap. 298, 477-484, 2001). TS-overexpressing, 5-FUdR-resistant HeLa cells have been established in order to examine resistance mechanisms and cross-resistance to 5-FU and raltitrexed. Treatment of 5-FUdR-resistant HeLa cells with TS antisense ODN effectively reduces TS mRNA and protein levels, and decreases the IC50 of 5-FUdR by up to 80% (Ferguson et al., Br. J. Pharmacol., 134, 1437-1446, 2001). These results indicate that antisense ODN treatment improves the efficacy of anti-TS chemotherapeutic drugs in vitro and in vivo, and is effective in overcoming tumor cell resistance to these drugs. However, cellular responses to antisense targeting of different TS mRNA domains are complex. In fact, targeting the translation start site (but not other TS mRNA regions) stimulates TS gene transcription (DeMoor et al., E.xp. Cell Res., 243, 11-21, 1998). Distinctive cellular responses to targeting of specific TS mRNA regions provide exciting therapeutic opportunities. Antisense ODN treatment to modulate TS activity, in combination with TS-targeting chemotherapeutic drugs, has the potential to be an effective anti-tumor therapy.


Assuntos
Antineoplásicos/farmacologia , RNA Mensageiro/genética , Timidilato Sintase/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Resistência a Medicamentos/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , RNA Antissenso/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/química , Timidilato Sintase/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
7.
Structure ; 6(7): 839-48, 1998 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9687366

RESUMO

BACKGROUND: Enzymes have evolved to recognise their target substrates with exquisite selectivity and specificity. Whether fragments of the substrate--perhaps never available to the evolving enzyme--are bound in the same manner as the parent substrate addresses the fundamental basis of specificity. An understanding of the relative contributions of individual portions of ligand molecules to the enzyme-binding interaction may offer considerable insight into the principles of substrate recognition. RESULTS: We report 12 crystal structures of Escherichia coli thymidylate synthase in complexes with available fragments of the substrate (dUMP), both with and without the presence of a cofactor analogue. The structures display considerable fidelity of binding mode and interactions. These complexes reveal several interesting features: the cofactor analogue enhances the localisation of substrate and substrate fragments near the reactive thiol; the ribose moiety reduces local disorder through additional specific enzyme-ligand interactions; the pyrimidine has multiple roles, ranging from stereospecificity to mechanistic competence; and the glycosidic linkage has an important role in the formation of a covalent attachment between substrate and enzyme. CONCLUSIONS: The requirements of ligand-protein binding can be understood in terms of the binding of separate fragments of the ligand. Fragments which are subsystems of the natural substrate for the enzyme confer specific contributions to the binding affinity, orientation or electrostatics of the enzymatic mechanism. This ligand-binding analysis provides a complementary method to the more prevalent approaches utilising site-directed mutagenesis. In addition, these observations suggest a modular approach for rational drug design utilising chemical fragments.


Assuntos
Nucleotídeos de Desoxiuracil/metabolismo , Timidilato Sintase/química , Timidilato Sintase/metabolismo , Cristalografia por Raios X , Nucleotídeos de Desoxiuracil/química , Desoxiuridina/química , Desoxiuridina/metabolismo , Escherichia coli/enzimologia , Ácido Fólico/análogos & derivados , Ácido Fólico/química , Ácido Fólico/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fosfatos/química , Fosfatos/metabolismo , Conformação Proteica , Quinazolinas/química , Quinazolinas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribose/química , Ribose/metabolismo , Relação Estrutura-Atividade , Timidilato Sintase/genética , Uridina/química , Uridina/metabolismo
8.
Protein Eng ; 6(7): 723-32, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7504257

RESUMO

We have added a chemical filter to the ligand placement algorithm of the molecular docking program DOCK. DOCK places ligands in receptors using local shape features. Here we label these shape features by chemical type and insist on complementary matches. We find fewer physically unrealistic complexes without reducing the number of complexes resembling the known ligand-receptor configurations. Approximately 10-fold fewer complexes are calculated and the new algorithm is correspondingly 10-fold faster than the previous shape-only matching. We tested the new algorithm's ability to reproduce three known ligand-receptor complexes: methotrexate in dihydrofolate reductase, deoxyuridine monophosphate in thymidylate synthase and pancreatic trypsin inhibitor in trypsin. The program found configurations within 1 A of the crystallographic mode, with fewer non-native solutions compared with shape-only matching. We also tested the program's ability to retrieve known inhibitors of thymidylate synthase and dihydrofolate reductase by screening molecular databases against the enzyme structures. Both algorithms retrieved many known inhibitors preferentially to other compounds in the database. The chemical matching algorithm generally ranks known inhibitors better than does matching based on shape alone.


Assuntos
Desenho de Fármacos , Ligantes , Tetra-Hidrofolato Desidrogenase/química , Timidilato Sintase/química , Tripsina/química , Algoritmos , Aprotinina/química , Aprotinina/metabolismo , Sítios de Ligação , Fenômenos Químicos , Físico-Química , Simulação por Computador , Cristalização , Nucleotídeos de Desoxiuracil/química , Nucleotídeos de Desoxiuracil/metabolismo , Eletroquímica , Metotrexato/química , Metotrexato/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Timidilato Sintase/metabolismo , Tripsina/metabolismo
9.
Gan To Kagaku Ryoho ; 19(7): 954-62, 1992 Jul.
Artigo em Japonês | MEDLINE | ID: mdl-1626951

RESUMO

Leucovorin (LV), given intravenously the orally becomes 5, 10-methylene tetrahydrofolate in both cancer and normal cells. FdUMP which is an active metabolite of 5-FU binds tightly to thymidylate synthase in the presence of the cofactor 5, 10-methylene tetrahydrofolate. This interaction leads to potentiate the cytotoxic effect of 5-FU by prolonged inhibition of thymidylate synthase. Clinically, the combination of LV and 5-FU is given parenterally by two schedules; 5 consecutive days schedule and weekly schedule. Five 5 consecutive days-schedule is divided into 2 methods. One is a 200 mg/m2/day of LV by Machover, and the other is 20 mg/m2/day of LV by O'Connell. The weekly schedule is a 2-hour infusion of dl-LV (500 mg/m2) and iv bolus of 5-FU (600 mg/m2), given 1 hour after the beginning of LV infusion by Petrelli. A multicenter cooperative study in Japan was conducted to evaluate the clinical efficacy of LV and 5-FU using the weekly schedule by Petrelli. Response rates were 31.5% and 41.2% against advanced gastric and colorectal cancer respectively. Then, we carried out a randomized early phase II study using 250 mg/m2 of l-LV weekly (similar to the schedule of Petrelli's, armA) and 100 mg/m2 (similar to the schedule of Machover's, arm B) or 10 mg/m2 (similar to the schedule of O'Connell's, arm C) of l-LV for 5 consecutive days against gastric cancer. The response rate was 33.3% in arm A, 24.1% in arm B and no response in arm C. Toxicity was within acceptable limits, Toxic effects included diarrhea, stomatitis, anorexia and myelohypoplasia. Our data suggests that high-dose LV and 5-FU seems to be a very promising combination but, there was no responder using low dose (10 mg/m2) of l-LV schedule against gastric cancer patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Avaliação de Medicamentos , Fluordesoxiuridilato/metabolismo , Fluoruracila/administração & dosagem , Fluoruracila/metabolismo , Humanos , Leucovorina/administração & dosagem , Leucovorina/farmacocinética , Timidilato Sintase/química , Timidilato Sintase/metabolismo
10.
Biochemistry ; 30(46): 11073-80, 1991 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-1932030

RESUMO

Replacement of methionine (Met) residues by selenomethionine (SeMet) was recently shown to facilitate the crystallographic analysis of protein structure through the application of multi-wavelength anomalous diffraction techniques [Yang et al. (1990) Science (Washington, D.C.) 249, 1398-1405]. The availability of SeMet-containing proteins provides an excellent opportunity to evaluate the effects of the complete replacement of Met by SeMet. We chose to compare the properties of selenomethionyl thymidylate synthase isolated from Escherichia coli DL41 (a methionine auxotroph) and wild-type (wt) enzyme obtained from E. coli Rue10. An improved purification procedure for thymidylate synthase was developed which permitted the isolation of 25 mg of pure protein from 2 g of E. coli in 90% yield in no more than 8 h. The pure wt and SeMet enzymes exhibited specific activities 40% higher than published values. Thermal stability studies at 30 degrees C in degassed buffer showed that the SeMet enzyme (t1/2 67 h) was 8-fold less stable than wt enzyme (t1/2 557 h). The half-lives for the latter enzymes in nondegassed buffers at 30 degrees C were decreased by 2-fold, thus indicating the sensitivity of the enzyme to dissolved oxygen. Both enzymes exhibited essentially the same kinetic and binding properties, including Km(dUMP) (1.2 x 10(-6) M), specificity constant (1.6 x 10(6) s-1 M-1), and Kd for 5-fluorodeoxyuridylate binding (1.2 nM) in covalent inhibitory ternary complexes. In addition, X-ray crystallographic analysis by difference Fourier synthesis showed there was no significant difference in conformation between the SeMet enzyme and the wt enzyme.


Assuntos
Escherichia coli/enzimologia , Selenometionina/análogos & derivados , Timidilato Sintase/isolamento & purificação , Sequência de Aminoácidos , Aminoácidos/química , Sítios de Ligação , Soluções Tampão , Estabilidade Enzimática , Escherichia coli/crescimento & desenvolvimento , Concentração de Íons de Hidrogênio , Cinética , Conformação Proteica , Selênio/química , Temperatura , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/química , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA