Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Environ Sci Pollut Res Int ; 31(1): 357-370, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38012491

RESUMEN

This study presents a comparative examination of the performance of green investment portfolios to attain a zero-carbon environment by studying assests-backed securities. Using the portfolio return, the draw ratio, or the ROMAD as measures, the numerical findings. The study intention is to measure and assess the success of green investment portfolios. This will probably include weighing the costs and advantages of several routes toward a carbon-free future, and the major emphasis is on measuring the effectiveness of financial investments in promoting ecological sustainability. The research specifically examines the use of digital finance and asset-backed securities (ABS). This research evaluates the efficacy of these two methodologies in promoting sustainable investments, using relevant scholarly literature and empirical evidence. The study examines many essential indicators, including risk-adjusted returns, carbon footprint reduction, and market stability. The research elucidates the merits and drawbacks of various approaches in fostering ecologically sustainable investments, drawing on case studies and real-world illustrations. The results of this study provide valuable insights into how digital finance and asset-backed securities (ABS) might effectively contribute to the attainment of zero-carbon objectives, all the while maintaining financial stability. This study provides valuable insights for politicians, investors, and financial institutions to make well-informed choices on implementing these initiatives in the context of sustainable development.


Asunto(s)
Carbono , Inversiones en Salud , Costos y Análisis de Costo , Huella de Carbono , Desarrollo Económico , China
2.
Biochemistry ; 51(38): 7551-68, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22928782

RESUMEN

We examined the cathepsin C-catalyzed hydrolysis of dipeptide substrates of the form Yaa-Xaa-AMC, using steady-state and pre-steady-state kinetic methods. The substrates group into three kinetic profiles based upon the broad range observed for k(cat)/K(a) and k(cat) values, pre-steady-state time courses, and solvent kinetic isotope effects (sKIEs). The dipeptide substrate Gly-Arg-AMC displayed large values for k(cat)/K(a) (1.6 ± 0.09 µM(-1) s(-1)) and k(cat) (255 ± 6 s(-1)), an inverse sKIE on k(cat)/K(a) ((D)(k(cat)/K(a)) = 0.6 ± 0.15), a modest, normal sKIE on k(cat) ((D)k(cat) = 1.6 ± 0.2), and immeasurable pre-steady-state kinetics, indicating an extremely fast pre-steady-state rate (>400 s(-1)). (Errors on fitted values are omitted in the text for clarity but may be found in Table 2.) These results conformed to a kinetic model where the acylation (k(ac)) and deacylation (k(dac)) half-reactions are very fast and similar in value. The second substrate type, Gly-Tyr-AMC and Ser-Tyr-AMC, the latter the subject of a comprehensive kinetic study (Schneck et al. (2008) Biochemistry 47, 8697-8710), were found to be less active substrates compared to Gly-Arg-AMC, with respective k(cat)/K(a) values of 0.49 ± 0.07 µM(-1 )s(-1) and 5.3 ± 0.5 µM(-1 )s(-1), and k(cat) values of 28 ± 1 s(-1) and 25 ± 0.5 s(-1). Solvent kinetic isotope effects for Ser-Tyr-AMC were found to be inverse for k(cat)/K(a) ((D)(k(cat)/K(a)) = 0.74 ± 0.05) and normal for k(cat) ((D)k(cat) = 2.3 ± 0.1) but unlike Gly-Arg-AMC, pre-steady-state kinetics of Gly-Tyr-AMC and Ser-Tyr-AMC were measurable and characterized by a single-exponential burst, with fast transient rates (490 s(-1) and 390 s(-1), respectively), from which it was determined that k(ac) ≫ k(dac) ∼ k(cat). The third substrate type, Gly-Ile-AMC, gave very low values of k(cat)/K(a) (0.0015 ± 0.0001 µM(-1) s(-1)) and k(cat) (0.33 ± 0.02 s(-1)), no sKIEs, ((D)(k(cat)/K(a)) = 1.05 ± 0.5 and (D)k(cat) = 1.06 ± 0.4), and pre-steady-state kinetics exhibited a discernible, but negligible, transient phase. For this third class of substrate, kinetic modeling was consistent with a mechanism in which k(dac) > k(ac) ∼ k(cat), and for which an isotope-insensitive step in the acylation half-reaction is the slowest. The combined results of these studies suggested that the identity of the amino acid at the P(1) position of the substrate is the main determinant of catalysis. On the basis of these kinetic data, together with crystallographic studies of substrate analogues and molecular dynamics analysis with models of acyl-enzyme intermediates, we present a catalytic model derived from the relative rates of the acylation vs deacylation half-reactions of cathepsin C. The chemical steps of catalysis are proposed to be dependent upon the conformational freedom of the amino acid substituents for optimal alignment for thiolation (acylation) or hydrolysis (deacylation). These studies suggest ideas for inhibitor design for papain-family cysteine proteases and strategies to progress drug discovery for other classes of disease-relevant cysteine proteases.


Asunto(s)
Aminoácidos/química , Catepsina C/química , Dipéptidos/química , Catálisis , Catepsina C/genética , Humanos , Concentración de Iones de Hidrógeno , Cinética , Simulación de Dinámica Molecular , Especificidad por Sustrato
3.
Nat Chem Biol ; 5(9): 647-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19648931

RESUMEN

Biochemical combinatorial techniques such as phage display, RNA display and oligonucleotide aptamers have proven to be reliable methods for generation of ligands to protein targets. Adapting these techniques to small synthetic molecules has been a long-sought goal. We report the synthesis and interrogation of an 800-million-member DNA-encoded library in which small molecules are covalently attached to an encoding oligonucleotide. The library was assembled by a combination of chemical and enzymatic synthesis, and interrogated by affinity selection. We describe methods for the selection and deconvolution of the chemical display library, and the discovery of inhibitors for two enzymes: Aurora A kinase and p38 MAP kinase.


Asunto(s)
ADN/química , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/síntesis química , Bibliotecas de Moléculas Pequeñas/síntesis química , Animales , Aurora Quinasas , Técnicas Químicas Combinatorias , ADN/genética , Modelos Moleculares , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
4.
Biochemistry ; 49(33): 7151-63, 2010 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-20597513

RESUMEN

Steady-state kinetic analysis of focal adhesion kinase-1 (FAK1) was performed using radiometric measurement of phosphorylation of a synthetic peptide substrate (Ac-RRRRRRSETDDYAEIID-NH(2), FAK-tide) which corresponds to the sequence of an autophosphorylation site in FAK1. Initial velocity studies were consistent with a sequential kinetic mechanism, for which apparent kinetic values k(cat) (0.052 +/- 0.001 s(-1)), K(MgATP) (1.2 +/- 0.1 microM), K(iMgATP) (1.3 +/- 0.2 microM), K(FAK-tide) (5.6 +/- 0.4 microM), and K(iFAK-tide) (6.1 +/- 1.1 microM) were obtained. Product and dead-end inhibition data indicated that enzymatic phosphorylation of FAK-tide by FAK1 was best described by a random bi bi kinetic mechanism, for which both E-MgADP-FAK-tide and E-MgATP-P-FAK-tide dead-end complexes form. FAK1 catalyzed the betagamma-bridge:beta-nonbridge positional oxygen exchange of [gamma-(18)O(4)]ATP in the presence of 1 mM [gamma-(18)O(4)]ATP and 1.5 mM FAK-tide with a progressive time course which was commensurate with catalysis, resulting in a rate of exchange to catalysis of k(x)/k(cat) = 0.14 +/- 0.01. These results indicate that phosphoryl transfer is reversible and that a slow kinetic step follows formation of the E-MgADP-P-FAK-tide complex. Further kinetic studies performed in the presence of the microscopic viscosogen sucrose revealed that solvent viscosity had no effect on k(cat)/K(FAK-tide), while k(cat) and k(cat)/K(MgATP) were both decreased linearly at increasing solvent viscosity. Crystallographic characterization of inactive versus AMP-PNP-liganded structures of FAK1 showed that a large conformational motion of the activation loop upon ATP binding may be an essential step during catalysis and would explain the viscosity effect observed on k(cat)/K(m) for MgATP but not on k(cat)/K(m) for FAK-tide. From the positional isotope exchange, viscosity, and structural data it may be concluded that enzyme turnover (k(cat)) is rate-limited by both reversible phosphoryl group transfer (k(forward) approximately 0.2 s(-1) and k(reverse) approximately 0.04 s(-1)) and a slow step (k(conf) approximately 0.1 s(-1)) which is probably the opening of the activation loop after phosphoryl group transfer but preceding product release.


Asunto(s)
Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/metabolismo , Péptidos/metabolismo , Adenosina Trifosfato/metabolismo , Adenilil Imidodifosfato/química , Adenilil Imidodifosfato/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Cinética , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Fosforilación , Unión Proteica
5.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 27(5): 489-92, 2010 Oct.
Artículo en Zh | MEDLINE | ID: mdl-20931522

RESUMEN

OBJECTIVE: To investigate the transforming growth factor beta induced (TGFBI; BIGH3) gene mutation and founder effect of two large Chinese families clinically diagnosed as Thiel-Behnke corneal dystrophy. METHODS: Fifteen members including 13 affected and 2 healthy in family A, 14 members including 6 affected and 8 healthy in family B, as well as 20 other unrelated healthy individuals were tested for TGFBI gene mutation. Haplotype analysis and clinical examination were also carried out in the two families. RESULTS: In exon 12 of the TGFBI gene, 1664G to A change was detected in all the patients, which leads to an amino acid replacement of arginine with glutamine (p.Arg555Gln). Members of the two families share some similar haplotypes. CONCLUSION: Genetic analysis is helpful in the diagnosis of corneal dystrophy. The two families may come from a same ancestor.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Efecto Fundador , Factor de Crecimiento Transformador beta/genética , Adolescente , Adulto , Anciano , Pueblo Asiatico/genética , Secuencia de Bases , Niño , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/genética , Exones , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Mutación Puntual , Adulto Joven
6.
Bioorg Med Chem Lett ; 19(15): 4441-5, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19497745

RESUMEN

The lead serum and glucocorticoid-related kinase 1 (SGK1) inhibitors 4-(5-phenyl-1H-pyrrolo[2,3-b]pyridin-3-yl)benzoic acid (1) and {4-[5-(2-naphthalenyl)-1H-pyrrolo[2,3-b]pyridin-3-yl]phenyl}acetic acid (2) suffer from low DNAUC values in rat, due in part to formation and excretion of glucuronic acid conjugates. These PK/glucuronidation issues were addressed either by incorporating a substituent on the 3-phenyl ring ortho to the key carboxylate functionality of 1 or by substituting on the group in between the carboxylate and phenyl ring of 2. Three of these analogs have been identified as having good SGK1 inhibition potency and have DNAUC values suitable for in vivo testing.


Asunto(s)
Química Farmacéutica/métodos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Diseño de Fármacos , Glucocorticoides/química , Ácido Glucurónico/química , Proteínas Inmediatas-Precoces/química , Concentración 50 Inhibidora , Modelos Químicos , Conformación Molecular , Inhibidores de Proteínas Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Ratas , Relación Estructura-Actividad
7.
Protein Sci ; 16(12): 2761-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17965184

RESUMEN

Serum and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine protein kinase of the AGC family which participates in the control of epithelial ion transport and is implicated in proliferation and apoptosis. We report here the 1.9 A crystal structure of the catalytic domain of inactive human SGK1 in complex with AMP-PNP. SGK1 exists as a dimer formed by two intermolecular disulfide bonds between Cys258 in the activation loop and Cys193. Although most of the SGK1 structure closely resembles the common protein kinase fold, the structure around the active site is unique when compared to most protein kinases. The alphaC helix is not present in this inactive form of SGK1 crystal structure; instead, the segment corresponding to the C helix forms a beta-strand that is stabilized by the N-terminal segment of the activation loop through a short antiparallel beta-sheet. Since the differences from other kinases occur around the ATP binding site, this structure can provide valuable insight into the design of selective and highly potent ATP-competitive inhibitors of SGK1 kinase.


Asunto(s)
Adenilil Imidodifosfato/química , Proteínas Inmediatas-Precoces/química , Proteínas Serina-Treonina Quinasas/química , Adenosina Trifosfato/metabolismo , Adenilil Imidodifosfato/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Dimerización , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Inmediatas-Precoces/aislamiento & purificación , Proteínas Inmediatas-Precoces/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Conformación Proteica , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
8.
J Med Chem ; 49(5): 1597-612, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16509577

RESUMEN

The syntheses, in vitro characterizations, and rat and monkey in vivo pharmacokinetic profiles of a series of 5-, 6-, and 7-methyl-substituted azepanone-based cathepsin K inhibitors are described. Depending on the particular regiochemical substitution and stereochemical configuration, methyl-substituted azepanones were identified that had widely varied cathepsin K inhibitory potency as well as pharmacokinetic properties compared to the 4S-parent azepanone analogue, 1 (human cathepsin K, K(i,app) = 0.16 nM, rat oral bioavailability = 42%, rat in vivo clearance = 49.2 mL/min/kg). Of particular note, the 4S-7-cis-methylazepanone analogue, 10, had a K(i,app) = 0.041 nM vs human cathepsin K and 89% oral bioavailability and an in vivo clearance rate of 19.5 mL/min/kg in the rat. Hypotheses that rationalize some of the observed characteristics of these closely related analogues have been made using X-ray crystallography and conformational analysis. These examples demonstrate the potential for modulation of pharmacological properties of cathepsin inhibitors by substituting the azepanone core. The high potency for inhibition of cathepsin K coupled with the favorable rat and monkey pharmacokinetic characteristics of compound 10, also known as SB-462795 or relacatib, has made it the subject of considerable in vivo evaluation for safety and efficacy as an inhibitor of excessive bone resorption in rat, monkey, and human studies, which will be reported elsewhere.


Asunto(s)
Azepinas/síntesis química , Conservadores de la Densidad Ósea/síntesis química , Catepsinas/antagonistas & inhibidores , Sulfonas/síntesis química , Animales , Azepinas/química , Azepinas/farmacología , Disponibilidad Biológica , Proteínas Sanguíneas/metabolismo , Conservadores de la Densidad Ósea/química , Conservadores de la Densidad Ósea/farmacología , Catepsina K , Catepsinas/química , Línea Celular , Permeabilidad de la Membrana Celular , Cristalografía por Rayos X , Haplorrinos , Humanos , Conformación Molecular , Unión Proteica , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Sulfonas/química , Sulfonas/farmacología
9.
ACS Med Chem Lett ; 7(3): 217-22, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26985301

RESUMEN

A novel series of potent and selective hexokinase 2 (HK2) inhibitors, 2,6-disubstituted glucosamines, has been identified based on HTS hits, exemplified by compound 1. Inhibitor-bound crystal structures revealed that the HK2 enzyme could adopt an "induced-fit" conformation. The SAR study led to the identification of potent HK2 inhibitors, such as compound 34 with greater than 100-fold selectivity over HK1. Compound 25 inhibits in situ glycolysis in a UM-UC-3 bladder tumor cell line via (13)CNMR measurement of [3-(13)C]lactate produced from [1,6-(13)C2]glucose added to the cell culture.

10.
Mol Cancer Ther ; 3(10): 1221-7, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15486189

RESUMEN

Inhibitors of the G(2) DNA damage checkpoint can selectively sensitize cancer cells with mutated p53 to killing by DNA-damaging agents. Isogranulatimide is a G(2) checkpoint inhibitor containing a unique indole/maleimide/imidazole skeleton identified in a phenotypic cell-based screen; however, the mechanism of action of isogranulatimide is unknown. Using natural and synthetic isogranulatimide analogues, we show that the imide nitrogen and a basic nitrogen at position 14 or 15 in the imidazole ring are important for checkpoint inhibition. Isogranulatimide shows structural resemblance to the aglycon of UCN-01, a potent bisindolemaleimide inhibitor of protein kinase C beta (IC(50), 0.001 micromol/L) and of the checkpoint kinase Chk1 (IC(50), 0.007 micromol/L). In vitro kinase assays show that isogranulatimide inhibits Chk1 (IC(50), 0.1 micromol/L) but not protein kinase C beta. Of 13 additional protein kinases tested, isogranulatimide significantly inhibits only glycogen synthase kinase-3beta (IC(50), 0.5 micromol/L). We determined the crystal structure of the Chk1 catalytic domain complexed with isogranulatimide. Like UCN-01, isogranulatimide binds in the ATP-binding pocket of Chk1 and hydrogen bonds with the backbone carbonyl oxygen of Glu(85) and the amide nitrogen of Cys(87). Unlike UCN-01, the basic N15 of isogranulatimide interacts with Glu(17), causing a conformation change in the kinase glycine-rich loop that may contribute importantly to inhibition. The mechanism by which isogranulatimide inhibits Chk1 and its favorable kinase selectivity profile make it a promising candidate for modulating checkpoint responses in tumors for therapeutic benefit.


Asunto(s)
Antineoplásicos/farmacología , Daño del ADN , Fase G2 , Imidazoles/farmacología , Indoles/farmacología , Proteínas Quinasas/metabolismo , Proteínas Quinasas/fisiología , Dominio Catalítico , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cristalografía por Rayos X , Cisteína/química , Relación Dosis-Respuesta a Droga , Glutamina/química , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Indoles/antagonistas & inhibidores , Concentración 50 Inhibidora , Maleimidas/antagonistas & inhibidores , Modelos Químicos , Modelos Moleculares , Nitrógeno/química , Fenotipo , Unión Proteica , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Estructura Terciaria de Proteína , Factores de Tiempo
11.
ACS Med Chem Lett ; 2(2): 142-7, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-24900293

RESUMEN

The discovery of potent and selective cyanamide-based inhibitors of the cysteine protease cathepsin C is detailed. Optimization of the template with regard to plasma stability led to the identification of compound 17, a potent cathepsin C inhibitor with excellent selectivity over other cathepsins and potent in vivo activity in a cigarette smoke mouse model.

12.
Protein Sci ; 17(10): 1791-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18662907

RESUMEN

VX-680, also known as MK-0457, is an ATP-competitive small molecule inhibitor of the Aurora kinases that has entered phase II clinical trials for the treatment of cancer. We have solved the cocrystal structure of AurA/TPX2/VX-680 at 2.3 A resolution. In the crystal structure, VX-680 binds to the active conformation of AurA. The glycine-rich loop in AurA adopts a unique bent conformation, forming a pi-pi interaction with the phenyl group of VX-680. In contrast, in the published AurA/VX-680 structure, VX-680 binds to AurA in the inactive conformation, interacting with a hydrophobic pocket only present in the inactive conformation. These data suggest that TPX2, a protein cofactor, can alter the binding mode of VX-680 with AurA. More generally, the presence of physiologically relevant cofactor proteins can alter the kinetics, binding interactions, and inhibition of enzymes, and studies with these multiprotein complexes may be beneficial to the discovery and optimization of enzyme inhibitors as therapeutic agents.


Asunto(s)
Proteínas de Ciclo Celular/química , Proteínas Asociadas a Microtúbulos/química , Proteínas Nucleares/química , Piperazinas/química , Proteínas Serina-Treonina Quinasas/química , Proteínas Recombinantes/química , Aurora Quinasas , Dominio Catalítico , Proteínas de Ciclo Celular/metabolismo , Cristalografía , Cristalografía por Rayos X , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Piperazinas/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/metabolismo
13.
Bioorg Med Chem Lett ; 13(6): 1191-4, 2003 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-12643941

RESUMEN

The design, synthesis and SAR of a series of 2,6,9-trisubstituted purine inhibitors of p38alpha kinase is reported. Synthetic routes were devised to allow for array synthesis in which all three points of diversity could be facilely explored. The binding of this novel series to p38alpha kinase, which was predicted to have several key interactions in common with SB-203580, was confirmed by X-ray crystallography of 19 (p38 IC(50)=82 nM).


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Urea/síntesis química , Urea/farmacología , Cristalografía por Rayos X , Diseño de Fármacos , Indicadores y Reactivos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Molecular , Relación Estructura-Actividad , Urea/análogos & derivados , Proteínas Quinasas p38 Activadas por Mitógenos
14.
J Biol Chem ; 277(48): 46609-15, 2002 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-12244092

RESUMEN

Chk1 is a serine-threonine kinase that plays an important role in the DNA damage response, including G(2)/M cell cycle control. UCN-01 (7-hydroxystaurosporine), currently in clinical trials, has recently been shown to be a potent Chk1 inhibitor that abrogates the G(2)/M checkpoint induced by DNA-damaging agents. To understand the structural basis of Chk1 inhibition by UCN-01, we determined the crystal structure of the Chk1 kinase domain in complex with UCN-01. Chk1 structures with staurosporine and its analog SB-218078 were also determined. All three compounds bind in the ATP-binding pocket of Chk1, producing only slight changes in the protein conformation. Selectivity of UCN-01 toward Chk1 over cyclin-dependent kinases can be explained by the presence of a hydroxyl group in the lactam moiety interacting with the ATP-binding pocket. Hydrophobic interactions and hydrogen-bonding interactions were observed in the structures between UCN-01 and the Chk1 kinase domain. The high structural complementarity of these interactions is consistent with the potency and selectivity of UCN-01.


Asunto(s)
Alcaloides/farmacología , Inhibidores de Proteínas Quinasas , Alcaloides/química , Secuencia de Aminoácidos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Proteínas Quinasas/química , Estaurosporina/análogos & derivados , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA