Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Chem Biol ; 13(7): 785-792, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28553945

RESUMEN

S-Adenosyl-L-methionine (SAM) is an enzyme cofactor used in methyl transfer reactions and polyamine biosynthesis. The biosynthesis of SAM from ATP and L-methionine is performed by the methionine adenosyltransferase enzyme family (Mat; EC 2.5.1.6). Human methionine adenosyltransferase 2A (Mat2A), the extrahepatic isoform, is often deregulated in cancer. We identified a Mat2A inhibitor, PF-9366, that binds an allosteric site on Mat2A that overlaps with the binding site for the Mat2A regulator, Mat2B. Studies exploiting PF-9366 suggested a general mode of Mat2A allosteric regulation. Allosteric binding of PF-9366 or Mat2B altered the Mat2A active site, resulting in increased substrate affinity and decreased enzyme turnover. These data support a model whereby Mat2B functions as an inhibitor of Mat2A activity when methionine or SAM levels are high, yet functions as an activator of Mat2A when methionine or SAM levels are low. The ramification of Mat2A activity modulation in cancer cells is also described.


Asunto(s)
Metionina Adenosiltransferasa/antagonistas & inhibidores , Quinolinas/farmacología , S-Adenosilmetionina/metabolismo , Triazoles/farmacología , Sitio Alostérico/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Cinética , Metionina Adenosiltransferasa/aislamiento & purificación , Metionina Adenosiltransferasa/metabolismo , Quinolinas/química , Relación Estructura-Actividad , Triazoles/química
2.
Anal Biochem ; 414(2): 179-86, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21402045

RESUMEN

The PI3K/AKT signaling pathway has an important regulatory role in cancer cell growth and tumorigenesis. Signal transduction through this pathway requires the assembly and activation of PDK1 and AKT at the plasma membrane. On activation of the pathway, PDK1 and AKT1/2 translocate to the membrane and bind to phosphatidylinositol-(3,4,5)-trisphosphate (PIP(3)) through interaction with their pleckstrin-homology domains. A biochemical method was developed to measure the kinase activity of PDK1 and AKT1/2, utilizing nickel-chelating coated lipid vesicles as a way to mimic the membrane environment. The presence of these vesicles in the reaction buffer enhanced the specific activity of the His-tagged PDK1 (full-length, and the truncated kinase domain) and the activity of the full-length His-tagged AKT1 and AKT2 when assayed in a cascade-type reaction. This enhanced biochemical assay is also suitable for measuring the inhibition of PDK1 by several selective compounds from the carbonyl-4-amino-pyrrolopyrimidine (CAP) series. One of these inhibitors, PF-5168899, was further evaluated using a high content cell-based assay in the presence of CHO cells engineered with GFP-PDK1.


Asunto(s)
Adenina/análogos & derivados , Pruebas de Enzimas/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Transducción de Señal , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Adenina/química , Adenina/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Histidina/genética , Histidina/metabolismo , Humanos , Cinética , Oligopéptidos/genética , Oligopéptidos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Pirazinas/química , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
Bioorg Med Chem Lett ; 21(1): 584-7, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21084194

RESUMEN

A novel series of pyrrolopyrazole-based protein kinase C ß II inhibitors has been identified from high-throughput screening. Herein, we report our initial structure-activity relationship studies with a focus on optimizing compound ligand efficiency and physicochemical properties, which has led to potent inhibitors with good cell permeability.


Asunto(s)
Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Pirazoles/química , Ensayos Analíticos de Alto Rendimiento , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/síntesis química , Pirazoles/farmacología , Relación Estructura-Actividad
4.
ACS Med Chem Lett ; 11(6): 1175-1184, 2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32550998

RESUMEN

Two novel compounds were identified as Naa50 binders/inhibitors using DNA-encoded technology screening. Biophysical and biochemical data as well as cocrystal structures were obtained for both compounds (3a and 4a) to understand their mechanism of action. These data were also used to rationalize the binding affinity differences observed between the two compounds and a MLGP peptide-containing substrate. Cellular target engagement experiments further confirm the Naa50 binding of 4a and demonstrate its selectivity toward related enzymes (Naa10 and Naa60). Additional analogs of inhibitor 4a were also evaluated to study the binding mode observed in the cocrystal structures.

5.
Biochemistry ; 48(41): 9823-30, 2009 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-19743875

RESUMEN

Checkpoint kinase 1 (CHK1) is a key element in the DNA damage response pathway and plays a crucial role in the S-G(2)-phase checkpoint. Inhibiting CHK1 is a therapeutic strategy involving abrogation of the G2/M mitotic checkpoint defense of tumor cells toward lethal damage induced by DNA-directed chemotherapeutic agents. To date, most CHK1 inhibition approaches have involved targeting the ATP site of this kinase. In this study, we provide crystallographic and kinetic characterization of two small molecule inhibitors that bind to an allosteric site in the proximity of the CHK1 substrate site. Analysis of kinetic and biophysical data has led to the conclusion that these small molecule allosteric site inhibitors of CHK1 are reversible and are neither ATP- nor peptide substrate-competitive. K(i) values of 1.89 and 0.15 microM, respectively, have been determined for these compounds using a mixed inhibitor kinetic analysis. Cocrystal structures of the inhibitors bound to CHK1 reveal an allosteric site, unique to CHK1, located in the C-terminal domain and consisting of a shallow groove linked to a small hydrophobic pocket. The pocket displays induced fit characteristics in the presence of the two inhibitors. These findings establish the potential for the development of highly selective CHK1 inhibitors.


Asunto(s)
Proteínas Quinasas/metabolismo , Regulación Alostérica , Sitios de Unión , Dominio Catalítico , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Clonación Molecular , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Modelos Moleculares , Fragmentos de Péptidos/química , Conformación Proteica , Proteínas Quinasas/química , Proteínas Quinasas/genética , Resonancia por Plasmón de Superficie
6.
Mol Cancer Ther ; 7(8): 2394-404, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18723486

RESUMEN

Checkpoints are present in all phases of the cell cycle and are regarded as the gatekeepers maintaining the integrity of the genome. Many conventional agents used to treat cancer impart damage to the genome and activate cell cycle checkpoints. Many tumors are defective in the tumor suppressor p53 and therefore lack a functional G(1) checkpoint. In these tumors, however, the S-G(2) checkpoints remain intact and, in response to DNA damage, arrest cell cycle progression allowing time for DNA repair. Checkpoint kinase 1 (Chk1) is a key element in the DNA damage response pathway and plays a crucial role in the S-G(2)-phase checkpoints. Inhibiting Chk1 represents a therapeutic strategy for creating a "synthetic lethal" response by overriding the last checkpoint defense of tumor cells against the lethal damage induced by DNA-directed chemotherapeutic agents. Chk1 inhibition is consistent with emerging targeted therapies aiming to exploit molecular differences between normal and cancer cells. Adding a Chk1 inhibitor to DNA-damaging cytotoxic therapy selectively targets tumors with intrinsic checkpoint defects while minimizing toxicity in checkpoint-competent normal cells. PF-00477736 was identified as a potent, selective ATP-competitive small-molecule inhibitor that inhibits Chk1 with a K(i) of 0.49 nM. PF-00477736 abrogates cell cycle arrest induced by DNA damage and enhances cytotoxicity of clinically important chemotherapeutic agents, including gemcitabine and carboplatin. In xenografts, PF-00477736 enhanced the antitumor activity of gemcitabine in a dose-dependent manner. PF-00477736 combinations were well tolerated with no exacerbation of side effects commonly associated with cytotoxic agents.


Asunto(s)
Benzodiazepinonas/farmacología , Daño del ADN , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/efectos de los fármacos , Pirazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cromatografía Liquida , Desoxicitidina/análogos & derivados , Desoxicitidina/antagonistas & inhibidores , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Fosforilación , Ratas , Ratas Sprague-Dawley , Fase S/efectos de los fármacos , Espectrometría de Masas en Tándem , Gemcitabina
7.
J Med Chem ; 50(22): 5253-6, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17887663

RESUMEN

The cocrystal structure of a library hit was used to design a novel series of CHK1 inhibitors. The new series retained the critical hydrogen-bonding groups of the resorcinol moiety for binding but lacked the phenolic anilide moiety. The newly designed compounds exhibited similar enzymatic activity, while demonstrating increased cellular potency. Compound 10c, showing no single agent effect, potentiated the antiproliferative effect of Gemcitabine in both prostate and breast cancer cell lines.


Asunto(s)
Antineoplásicos/síntesis química , Compuestos de Bifenilo/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/metabolismo , Pirazoles/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Neoplasias de la Mama , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cristalografía por Rayos X , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Humanos , Indazoles/química , Masculino , Modelos Moleculares , Estructura Molecular , Neoplasias de la Próstata , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química , Pirazoles/química , Pirazoles/farmacología , Relación Estructura-Actividad , Gemcitabina
8.
J Biomol Screen ; 7(6): 531-40, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14599351

RESUMEN

Components within synthetic chemical and natural product extract libraries often interfere with fluorescence-based assays. Fluorescence interference can result when the intrinsic spectral properties of colored compounds overlap with the fluorescent probes. Typically, fluorescence-based protease assays use peptide amidomethylcoumarin derivatives as substrates. However, because many organic compounds absorb in the ultraviolet region, they can interfere with coumarin-based fluorescence assays. Red-shifted fluorescent dyes such as peptidyl rhodamine derivatives are useful because there is generally less interference from organic compounds outside the ultraviolet wavelengths. In this report, rhodamine-based fluorogenic substrates, such as bis-(Leu)(2)-Rhod110 and bis-(Ala-Pro)-Rhod110, were developed for leucine aminopeptidase and dipeptidyl aminopeptidase. Novel, tandem rhodamine substrates such as Ala-Pro-Rhod110-Leu were designed with 2 protease cleavage sites and used to assay 2 proteases in a multiplex format. General endpoint high-throughput screening (HTS) assays were also developed for leucine aminopeptidase, dipeptidyl aminopeptidase, and trypsin that incorporated both amidomethylcoumarin and rhodamine-based fluorogenic substrates into a single screening format. These dual-substrate assays allowed for the successful screening of the LOPAC trade mark collection and natural product extracts despite high levels of fluorescence interference.


Asunto(s)
Baclofeno/análogos & derivados , Bioensayo/métodos , Endopeptidasas/análisis , Colorantes Fluorescentes/química , Rodaminas/química , Baclofeno/análisis , Baclofeno/farmacología , Técnicas Químicas Combinatorias/métodos , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/antagonistas & inhibidores , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Endopeptidasas/metabolismo , Concentración 50 Inhibidora , Leucil Aminopeptidasa/metabolismo , Pentamidina/análisis , Pentamidina/farmacología , Péptidos/química , Péptidos/metabolismo , Inhibidores de Proteasas/análisis , Inhibidores de Proteasas/metabolismo , Especificidad por Sustrato
9.
Biosci Rep ; 34(2)2014 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27919031

RESUMEN

The PKN (protein kinase N) family of Ser/Thr protein kinases regulates a diverse set of cellular functions, such as cell migration and cytoskeletal organization. Inhibition of tumour PKN activity has been explored as an oncology therapeutic approach, with a PKN3-targeted RNAi (RNA interference)-derived therapeutic agent in Phase I clinical trials. To better understand this important family of kinases, we performed detailed enzymatic characterization, determining the kinetic mechanism and lipid sensitivity of each PKN isoform using full-length enzymes and synthetic peptide substrate. Steady-state kinetic analysis revealed that PKN1-3 follows a sequential ordered Bi-Bi kinetic mechanism, where peptide substrate binding is preceded by ATP binding. This kinetic mechanism was confirmed by additional kinetic studies for product inhibition and affinity of small molecule inhibitors. The known lipid effector, arachidonic acid, increased the catalytic efficiency of each isoform, mainly through an increase in kcat for PKN1 and PKN2, and a decrease in peptide KM for PKN3. In addition, a number of PKN inhibitors with various degrees of isoform selectivity, including potent (Ki<10 nM) and selective PKN3 inhibitors, were identified by testing commercial libraries of small molecule kinase inhibitors. This study provides a kinetic framework and useful chemical probes for understanding PKN biology and the discovery of isoform-selective PKN-targeted inhibitors.


Asunto(s)
Adenosina Trifosfato/química , Ácido Araquidónico/química , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/química , Inhibidores de Proteínas Quinasas/química , Catálisis , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Cinética
10.
ACS Chem Biol ; 8(5): 978-86, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23441851

RESUMEN

The contributions of the phosphoacceptor and the catalytic domain context to protein kinase biology and inhibitor potency are routinely overlooked, which can lead to mischaracterization of inhibitor and receptor functions. The receptor tyrosine kinase vascular endothelial growth factor receptor-2 (VEGFR2) is studied as a model system using a series of phosphoacceptor substrates (k(cat)/K(m) 684-116,000 M(-1) s(-1)) to assess effects on catalysis and inhibitor binding. ATP-competitive inhibitor potency toward the VEGFR2 catalytic domain (VEGFR2-CD) varies with different phosphoacceptor substrates, which is unexpected because the phosphoacceptors do not affect K(m,ATP) values. Indazole-based inhibitors are up to 60-fold more potent with two substrates (gastrin, minigastrin) relative to the others. Thus there is a component of uncompetitive inhibition because a specific phosphoacceptor enhances potency but is not strictly required. This substrate-specific inhibitory potency enhancement correlates with phosphoacceptor active site saturation and is not observed with other related kinases. The effect is confined to a specific catalytic domain conformation because autophosphorylation eliminates the potency enhancement as does the addition of the juxtamembrane domain (20 amino acids). Indazole inhibitor structure-activity analysis reveals that the magnitude of potency enhancement correlates with the size of the substituent that binds in a regulatory region of the active site. VEGFR drugs profiled with VEGFR2-CD using minigastrin have potency well-correlated with inhibition of full-length, cellular VEGFR2 autophosphorylation, an indication that the minigastrin-induced conformation is biologically relevant. These findings raise the possibility that inhibitors directed toward a common target can have different biological effects based on the kinase-substrate complexes present in different cellular contexts.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Caseínas/metabolismo , Dominio Catalítico , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Gastrinas/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular , Cinética , Péptidos/metabolismo , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Especificidad por Sustrato , Factor A de Crecimiento Endotelial Vascular/farmacología
11.
Chem Biol ; 18(1): 3-4, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21276931

RESUMEN

A major challenge for the discovery of protein kinase inhibitors is to identify potent, selective, and novel pharmacophores. In this issue, Fedorov et al. (2011) describes KH-CB19, an ATP-competitive inhibitor of cdc2-like kinase that interacts with the ATP hinge region through a halogen-bonding motif.

12.
Eur J Pharmacol ; 627(1-3): 16-25, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19850035

RESUMEN

Protein kinase C (PKC) family members such as PKCbetaII may become activated in the hyperglycemic state associated with diabetes. Preclinical and clinical data implicate aberrant PKC activity in the development of diabetic microvasculature abnormalities. Based on this potential etiological role for PKC in diabetic complications, several therapeutic PKC inhibitors have been investigated in clinical trials for the treatment of diabetic patients. In this report, we present the discovery and preclinical evaluation of a novel class of 3-amino-pyrrolo[3,4-c]pyrazole derivatives as inhibitors of PKC that are structurally distinct from the prototypical indolocarbazole and bisindolylmaleimide PKC inhibitors. From this pyrrolo-pyrazole series, several compounds were identified from biochemical assays as potent, ATP-competitive inhibitors of PKC activity with high specificity for PKC over other protein kinases. These compounds were also found to block PKC signaling activity in multiple cellular functional assays. PF-04577806, a representative from this series, inhibited PKC activity in retinal lysates from diabetic rats stimulated with phorbol myristate acetate. When orally administered, PF-04577806 showed good exposure in the retina of diabetic Long-Evans rats and ameliorated retinal vascular leakage in a streptozotocin-induced diabetic rat model. These novel PKC inhibitors represent a promising new class of targeted protein kinase inhibitors with potential as therapeutic agents for the treatment of patients with diabetic microvascular complications.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Descubrimiento de Drogas , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Enfermedades de la Retina/metabolismo , Vasos Retinianos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Administración Oral , Animales , Bovinos , Línea Celular , Complicaciones de la Diabetes/tratamiento farmacológico , Complicaciones de la Diabetes/enzimología , Modelos Animales de Enfermedad , Humanos , Masculino , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/administración & dosificación , Pirazoles/química , Pirazoles/farmacocinética , Pirazoles/farmacología , Ratas , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/enzimología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Especificidad por Sustrato
13.
Biochemistry ; 45(47): 13970-81, 2006 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-17115692

RESUMEN

The conventional protein kinase C isoform, PKCII, is a signaling kinase activated during the hyperglycemic state and has been associated with the development of microvascular abnormalities associated with diabetes. PKCII, therefore, has been identified as a therapeutic target where inhibitors of its kinase activity are being pursued for treatment of microvascular-related diabetic complications. In this report, we describe the crystal structure of the catalytic domain of PKCbetaII complexed with an inhibitor at 2.6 A resolution. The kinase domain of PKCbetaII was cleaved and purified from full-length PKCbetaII expressed in baculovirus-infected insect cells. The overall kinase domain structure follows the classical bilobal fold and is in its fully activated conformation with three well-defined phosphorylated residues: Thr-500, Thr-641, and Ser-660. Different from the crystal structures of nonconventional PKC isoforms, the C-terminus of the PKCbetaII catalytic domain is almost fully ordered and features a novel alpha helix in the turn motif. An ATP-competitive inhibitor, 2-methyl-1H-indol-3-yl-BIM-1, was crystallized with the PKCbetaII catalytic domain as a dimer of two enzyme-inhibitor complexes. The bound inhibitor adopts a nonplanar conformation in the ATP-binding site, with the kinase domain taking on an intermediate, open conformation. This PKCbetaII-inhibitor complex represents the first structural description of any conventional PKC kinase domain. Given the pathogenic role of PKCbetaII in the development of diabetic complications, this structure can serve as a template for the rational design of inhibitors as potential therapeutic agents.


Asunto(s)
Indoles/farmacología , Maleimidas/farmacología , Proteína Quinasa C/química , Inhibidores de Proteínas Quinasas/farmacología , Secuencia de Aminoácidos , Dominio Catalítico , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Fosforilación , Conformación Proteica , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C beta , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido
14.
Bioorg Med Chem Lett ; 15(8): 1997-2001, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15808455

RESUMEN

Syntheses and nitric oxide synthase inhibitory activity of cyclic amidines containing 5,6- 6,6- and 7,6-fused systems are described. X-ray structure determination facilitated the assignment of the stereochemistry of the most active compounds perhydro-2-iminoisoquinoline (8a) and perhydro-2-iminopyrindine (10a). Both 8a and 10a are very potent inhibitors of iNOS, with excellent selectivity over eNOS and they are orally active in rats with long duration suitable for once or twice a day dosing.


Asunto(s)
Amidinas/química , Compuestos Bicíclicos con Puentes/química , Inhibidores Enzimáticos/administración & dosificación , Óxido Nítrico Sintasa/antagonistas & inhibidores , Piridinas/química , Quinolinas/química , Administración Oral , Amidinas/administración & dosificación , Animales , Compuestos Bicíclicos con Puentes/administración & dosificación , Línea Celular , Inhibidores Enzimáticos/farmacología , Óxido Nítrico Sintasa de Tipo II , Piridinas/administración & dosificación , Quinolinas/administración & dosificación , Ratas
16.
Bioorg Med Chem Lett ; 14(17): 4539-44, 2004 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-15357988

RESUMEN

Syntheses and evaluation of pyrrolidin-2-imines and 1,3-thiazolidin-2-imines as inhibitors of nitric oxide synthase (NOS) are discussed. An extensive SAR was established for pyrrolidin-2-imines class of compounds. The amidines came out as the most potent inhibitors in addition to displaying selectivity.


Asunto(s)
Inhibidores Enzimáticos/química , Iminas/química , Óxido Nítrico Sintasa/antagonistas & inhibidores , Pirrolidinas/química , Tiazoles/química , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Iminas/farmacología , Óxido Nítrico Sintasa/metabolismo , Pirrolidinas/farmacología , Tiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA