Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 11436, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31391482

RESUMO

Proteases are one of attractive therapeutic targets to play key roles in pharmacological action. There are many protease inhibitors in nature, and most of them structurally have cystine knot motifs. Their structures are favorable for recognition of active pockets of proteases, leading to the potent inhibition. However, they also have drawbacks, such as broad cross-reactivity, on the therapeutic application. To create therapeutic proteins derived from a disulfide-rich scaffold, we selected human serine protease inhibitor Kazal type 2 (SPINK2) through a scaffold screening, as a protein scaffold with requirements for therapeutic proteins. We then constructed a diverse library of the engineered SPINK2 by introducing random mutations into its flexible loop region with the designed method. By phage panning against four serine proteases, we isolated potent inhibitors against each target with picomolar KD and sub-nanomolar Ki values. Also, they exhibited the desired specificities against target proteases without inhibiting non-target proteases. The crystal structure of kallikrein related peptidase 4 (KLK4)-engineered SPINK2 complex revealed the interface with extensive conformational complementarity. Our study demonstrates that engineered SPINK2 can serve as a scaffold to generate therapeutic molecules against target proteins with groove structures.


Assuntos
Desenho de Fármacos , Glicoproteínas/farmacologia , Mutagênese , Engenharia de Proteínas/métodos , Inibidores de Serinopeptidase do Tipo Kazal/farmacologia , Inibidores de Serina Proteinase/farmacologia , Cristalografia por Raios X , Glicoproteínas/genética , Glicoproteínas/uso terapêutico , Glicoproteínas/ultraestrutura , Calicreínas/metabolismo , Calicreínas/ultraestrutura , Modelos Moleculares , Estrutura Terciária de Proteína , Inibidores de Serinopeptidase do Tipo Kazal/genética , Inibidores de Serinopeptidase do Tipo Kazal/uso terapêutico , Inibidores de Serinopeptidase do Tipo Kazal/ultraestrutura , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/uso terapêutico , Relação Estrutura-Atividade
2.
Drug Metab Dispos ; 41(4): 878-87, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23378626

RESUMO

The absorption, metabolism, and excretion of (2R,3R,4R)-4-hydroxy-2-(hydroxymethyl)pyrrolidin-3-yl 4-O-(6-deoxy-ß-d-glucopyranosyl)-α-d-glucopyranoside (CS-1036), a novel and potent pancreatic and salivary α-amylase inhibitor, were evaluated in F344/DuCrlCrlj rats and cynomolgus monkeys. The total body clearance and volume of distribution of CS-1036 were low (2.67-3.44 ml/min/kg and 0.218-0.237 l/kg for rats and 2.25-2.84 ml/min/kg and 0.217-0.271 l/kg for monkeys). After intravenous administration of [(14)C]CS-1036 to rats and monkeys, radioactivity was mainly excreted into urine (77.2% for rats and 81.1% for monkeys). After oral administration, most of the radioactivity was recovered from feces (80.28% for rats and 88.13% for monkeys) with a low oral bioavailability (1.73-2.44% for rats and 0.983-1.20% for monkeys). In rats, intestinal secretion is suggested to be involved in the fecal excretion as a minor component because fecal excretion after intravenous administration was observed (15.66%) and biliary excretion was almost negligible. Although intestinal flora was involved in CS-1036 metabolism, CS-1036 was the main component in feces (70.3% for rats and 48.7% for monkeys) and in the intestinal contents (33-68% for rats up to 2 hours after the dose) after oral administration. In Zucker diabetic fatty rats, CS-1036 showed a suppressive effect on plasma glucose elevation after starch loading with a 50% effective dose at 0.015 mg/kg. In summary, CS-1036 showed optimal pharmacokinetic profiles: low oral absorption and favorable stability in gastrointestinal lumen, resulting in suppression of postprandial hyperglycemia by α-amylase inhibition.


Assuntos
Glicemia/efeitos dos fármacos , Dissacarídeos/farmacocinética , Absorção Intestinal/efeitos dos fármacos , Pirrolidinas/farmacocinética , alfa-Amilases/antagonistas & inibidores , Absorção , Administração Oral , Animais , Disponibilidade Biológica , Dissacarídeos/administração & dosagem , Dissacarídeos/farmacologia , Injeções Intravenosas , Macaca fascicularis , Masculino , Pirrolidinas/administração & dosagem , Pirrolidinas/farmacologia , Ratos , Distribuição Tecidual
3.
J Med Microbiol ; 61(Pt 9): 1280-1285, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22700553

RESUMO

PyrG (CTP synthase) catalyses the conversion of UTP to CTP, an essential step in the pyrimidine metabolic pathway in a variety of bacteria, including those causing community-acquired respiratory tract infections (RTIs). In this study, a luminescence-based ATPase assay of PyrG was developed and used to evaluate the inhibitory activity of 2-(3-[3-oxo-1,2-benzisothiazol-2(3H)-yl]phenylsulfonylamino) benzoic acid (compound G1). Compound G1 inhibited PyrG derived from Streptococcus pneumoniae with a 50 % inhibitory concentration value of 0.091 µM, and the inhibitory activity of compound G1 was 13 times higher than that of acivicin (1.2 µM), an established PyrG inhibitor. The results of saturation transfer difference analysis using nuclear magnetic resonance spectroscopy suggested that these compounds compete with ATP and/or UTP for binding to Strep. pneumoniae PyrG. Finally, compound G1 was shown to have antimicrobial activity against several different bacteria causing RTIs, such as Staphylococcus aureus and Haemophilus influenzae, suggesting that it is a prototype chemical compound that could be harnessed as an antimicrobial drug with a novel structure to target bacterial PyrG.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Carbono-Nitrogênio Ligases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Infecções Respiratórias/microbiologia , Adenosina Trifosfatases/metabolismo , Antibacterianos/química , Bactérias/enzimologia , Proteínas de Bactérias/antagonistas & inibidores , Ácido Benzoico/química , Ácido Benzoico/farmacologia , Citidina Trifosfato/metabolismo , Inibidores Enzimáticos/química , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/enzimologia , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana/métodos , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/enzimologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/enzimologia , Uridina Trifosfato/metabolismo
4.
Bioorg Med Chem Lett ; 22(9): 3083-8, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22487179

RESUMO

We have previously disclosed 1,2,4-oxadiazole derivative 3 as a potent S1P(3)-sparing S1P(1) agonist. Although compound 3 exhibits potent and manageable immunosuppressive efficacy in various in vivo models, recent studies have revealed that its 1,2,4-oxadiazole ring is subjected to enterobacterial decomposition. As provisions for unpredictable issues, a series of alternative compounds were synthesized on the basis of compound 3. Extensive SAR studies led to the finding of 1,3-thiazole 24c with the EC(50) value of 3.4 nM for human S1P(1), and over 5800-fold selectivity against S1P(3). In rat on host versus graft reaction (HvGR), the ID(50) value of 24c was determined at 0.07 mg/kg. The pharmacokinetics in rat and monkey is also reported. Compared to compound 3, 24c showed excellent stability against enterobacteria.


Assuntos
Piridinas/síntese química , Receptores de Lisoesfingolipídeo/agonistas , Tiazóis/química , Tiofenos/síntese química , Animais , Haplorrinos , Humanos , Piridinas/farmacologia , Ratos , Relação Estrutura-Atividade , Tiazóis/farmacologia , Tiofenos/farmacologia
5.
FEMS Microbiol Lett ; 330(2): 121-6, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22428584

RESUMO

PyrH is a member of the UMP kinase family that catalyses the conversion of UMP to UDP, an essential step in the pyrimidine metabolic pathway in a variety of bacteria including those causing community-acquired respiratory tract infections (RTIs). In this study, we have developed a luminescence-based kinase assay of PyrH and evaluated the inhibitory activity of PYRH-1 (sodium {3-[4-tert-butyl-3-(9H-xanthen-9-ylacetylamino)phenyl]-1-cyclohexylmethylpropoxycarbonyloxy}acetate). PYRH-1 inhibits PyrH derived from both Streptococcus pneumoniae and Haemophilus influenzae with IC(50) (concentration of inhibitor giving a 50% decrease in enzyme activity) values of 48 and 75 µM, respectively, whose inhibitory activity against S. pneumoniae PyrH was far higher compared with that of UTP (IC(50)  = 710 µM), an allosteric PyrH inhibitor. The molecular interaction analysis by surface plasmon resonance suggested that PYRH-1 directly interacts with S. pneumoniae PyrH at one-to-one molar ratio. Finally, PYRH-1 was shown to have antimicrobial activity against several different bacteria causing RTIs, such as S. pneumoniae, Staphylococcus aureus, H. influenzae (acrA knockout strain), suggesting that PYRH-1 is a prototype chemical compound that can be harnessed as an antimicrobial drug with a novel mode of action by targeting bacterial PyrH.


Assuntos
Inibidores Enzimáticos/isolamento & purificação , Haemophilus influenzae/enzimologia , Núcleosídeo-Fosfato Quinase/antagonistas & inibidores , Streptococcus pneumoniae/enzimologia , Inibidores Enzimáticos/metabolismo , Haemophilus influenzae/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana , Núcleosídeo-Fosfato Quinase/metabolismo , Ligação Proteica , Staphylococcus aureus/efeitos dos fármacos , Streptococcus pneumoniae/efeitos dos fármacos , Ressonância de Plasmônio de Superfície , Difosfato de Uridina/metabolismo , Uridina Monofosfato/metabolismo
6.
FEMS Microbiol Lett ; 331(2): 113-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22458538

RESUMO

Validation of bactericidal profiles owing to a deficiency of target bacterial molecule provides opportunities to discover antimicrobial drug candidates. In this study, we constructed genetic-engineered Escherichia coli strains, in which the target gene expression is conditionally regulated by a tryptophan promoter, while the target protein expression is regulated by N-end rule-based proteolysis. Among 10 genes, whose correspondent proteins are target candidates of antibiotics for community acquired respiratory tract infection, it was clearly demonstrated that the suppression of DnaB, GlmU, or DnaX results in a bactericidal profile, while the suppression of FabB, PyrG, DnaG, Der, PyrH, Era, or IspA leads to a bacteriostatic profile. This study is the first to predict the antibacterial inhibition profiles of Der, DnaG, DnaX, Era, GlmU, IspA, PyrG, and PyrH, and confirms previous findings for DnaB and FabB. The results suggested that the system constructed in this study is a novel and useful tool to validate whether the target bacterial molecule has appropriate properties as a target of antimicrobial agents.


Assuntos
Anti-Infecciosos/farmacologia , Proteínas de Bactérias/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Testes de Sensibilidade Microbiana/métodos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Genes Essenciais/efeitos dos fármacos , Genes Essenciais/fisiologia , Engenharia Genética/métodos , Humanos , Regiões Promotoras Genéticas , Proteólise , Infecções Respiratórias/microbiologia , Triptofano/farmacologia
7.
Antimicrob Agents Chemother ; 54(12): 5298-302, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20921311

RESUMO

Tomopenem (formerly CS-023) is a novel carbapenem with broad-spectrum activities against diverse hospital pathogens, including Pseudomonas aeruginosa and methicillin-resistant Staphylococcus aureus (MRSA). We examined the in vivo pharmacodynamic characteristics of tomopenem against P. aeruginosa and MRSA by using a neutropenic murine thigh infection model with P. aeruginosa 12467 (MIC, 1 µg/ml) and MRSA 12372 (MIC, 2 µg/ml). The mice had 10(6) to 10(7) CFU/thigh of each strain 2 h after inoculation and were treated for 24 h with a fractionated administration of tomopenem given at intervals of 3, 6, 12, and 24 h. The serum protein binding of tomopenem was 17.4%. The efficacy of tomopenem in both infection models was enhanced by frequent dosing, which indicates that the efficacy is driven by the time above MIC (T(MIC)). In a sigmoid model, the cumulative percentages of the 24-h period that the concentrations of free, unbound fractions of the drug exceeded the MIC under steady-state pharmacokinetic conditions (f%T(MIC)s) were best correlated with efficacy when R(2) was 0.79 and 0.86 against P. aeruginosa and MRSA, respectively. Other pharmacokinetic and pharmacodynamic (PK-PD) indexes for the free, unbound fractions, the area under the concentration-time curve over 24 h in the steady state divided by the MIC (AUC/MIC) and the maximum concentration of the drug in serum divided by the MIC (C(max)/MIC), showed poor correlation with efficacy when R(2) was ≤0.42. The f%T(MIC) values required for a static effect, 1-log kill, and 2-log kill against P. aeruginosa were 29, 39, and 51, respectively, which were similar to those for meropenem, for which the values were 24, 33, and 45, respectively. Against MRSA, the values for tomopenem were 27, 35, and 47. In conclusion, the pharmacodynamic characteristics of tomopenem were similar to those of meropenem against P. aeruginosa, and there was no difference between the target values for P. aeruginosa and MRSA required for efficacy in this study.


Assuntos
Antibacterianos , Carbapenêmicos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Animais , Antibacterianos/farmacocinética , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Carbapenêmicos/farmacocinética , Carbapenêmicos/farmacologia , Carbapenêmicos/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos ICR , Coxa da Perna/microbiologia
8.
Antimicrob Agents Chemother ; 53(1): 186-92, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18955520

RESUMO

Two neuraminidase (NA) inhibitors, zanamivir (Relenza) and oseltamivir phosphate (Tamiflu), have been licensed for the treatment of and prophylaxis against influenza. In this paper, the new potent NA inhibitor R-125489 is reported for the first time. R-125489 inhibited the NA activities of various type A and B influenza viruses, including subtypes N1 to N9 and oseltamivir-resistant viruses. The survival effect of R-125489 was shown to be similar to that of zanamivir when administered intranasally in a mouse influenza virus A/Puerto Rico/8/34 infection model. Moreover, we found that the esterified form of R-125489 showed improved efficacy compared to R-125489 and zanamivir, depending on the acyl chain length, and that 3-(O)-octanoyl R-125489 (CS-8958) was the best compound in terms of its life-prolonging effect (P < 0.0001, compared to zanamivir) in the same infection model. A prolonged survival effect was observed after a single administration of CS-8958, even if it was given 7 days before infection. It is suggested that intranasally administered CS-8958 works as a long-acting NA inhibitor and shows in vivo efficacy as a result of a single intranasal administration.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Neuraminidase/antagonistas & inibidores , Pró-Fármacos/farmacologia , Animais , Antivirais/administração & dosagem , Antivirais/química , Linhagem Celular , Cães , Células HeLa , Humanos , Camundongos , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Zanamivir/administração & dosagem , Zanamivir/farmacologia
9.
J Bacteriol ; 184(17): 4933-5, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12169621

RESUMO

The dif site is located in the replication terminus region of bacterial chromosomes, having a function of resolving dimeric chromosomes formed during replication. We demonstrate that filamentous bacteriophages of vibrios, such as f237 (Vibrio parahaemolyticus) and CTXphi (V. cholerae), are integrated into the dif-like site of host chromosome.


Assuntos
Cromossomos Bacterianos , Vibrio parahaemolyticus/virologia , Integração Viral , Vibrio parahaemolyticus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...