Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bioorg Med Chem ; 73: 117035, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36208543

RESUMEN

Hereditary angioedema (HAE) is a rare and potentially life-threatening disease that affects an estimated 1 in 50,000 individuals worldwide. Berotralstat (BCX7353) is the only small molecule approved by the US Food and Drug Administration (FDA) for the prophylactic treatment of HAE attacks in patients 12 years and older. During the discovery of BCX7353, we also identified a novel series of small molecules containing a quaternary carbon as potent and orally bioavailable Plasma Kallikrein (PKal) inhibitors. Lead compound was identified as a potent inhibitor following a detailed lead optimization process that balanced the lipophilic efficiency (LipE) and pharmacokinetic (PK) profile.


Asunto(s)
Angioedemas Hereditarios , Calicreína Plasmática , Angioedemas Hereditarios/tratamiento farmacológico , Angioedemas Hereditarios/prevención & control , Antivirales/uso terapéutico , Carbono , Humanos , Estados Unidos
2.
Bioorg Med Chem ; 74: 117034, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36272185

RESUMEN

The three complement pathways comprising the early phase of the complement system (the classical, lectin, and alternative pathways) act together with the innate and adaptive immune systems to protect against foreign entities and maintain tissue homeostasis. While these systems are normally under tight regulatory control, several diseases have been reported to correlate with uncontrolled activation and amplification of the alternative pathway, including paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, C3 glomerulopathy, and age-related macular degeneration. Complement FactorD (CFD), a serine protease, is the rate-limiting enzyme for the activity of alternative pathway. CFD activates the alternative pathway by cleaving Complement Factor B complexed to C3b (C3bB) to generate alternative pathway C3 convertase (C3bBb). In our search for novel CFD inhibitors with therapeutic potential, we employed a hot-spot analysis of an ensemble of apo and holo CFD structures. This analysis identified potential pharmacophore features that aided in the design of a series of compounds based on an l-proline core. While these compounds inhibited CFD in an esterolytic assay (for example, a proline-based compound, IC50 = 161 nM), the pharmacokinetic (PK) properties were poor. A strategy of scaffold hopping via ring opening led to a novel series of acyclic compounds, with subsequent structure-based ligand design and lead optimization producing several novel CFD inhibitors. One of these inhibitors, 1-(2-((2-(3-chloro-2-fluorobenzylamino)-2-oxoethyl)(cyclopropyl)amino)-2-oxoethyl)-5-(3-methyl-3-(1-methylpiperidin-4-yl)ureido)-1H-indazole-3-carboxamide, showed good potency with IC50s of 37 nM in the esterolytic assay and 30 nM in a hemolytic assay and PK assessments following oral administration to rats revealed a Cmax of 113 ng/mL and an AUC0-24h of 257 hr.ng/mL.


Asunto(s)
Factor D del Complemento , Serina Endopeptidasas , Ratas , Animales , Factor D del Complemento/metabolismo , Hemólisis , Ligandos
3.
J Biol Chem ; 292(38): 15907-15915, 2017 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-28794158

RESUMEN

Purine nucleoside phosphorylase (PNP) is part of the human purine salvage pathway. Its deficiency triggers apoptosis of activated T-cells, making it a target for T-cell proliferative disorders. Transition-state analogues of PNP bind with picomolar (pm) dissociation constants. Tight-binding PNP inhibitors show exceptionally long lifetimes on the target enzyme. We solve the mechanism of the target residence time by comparing functional off-rates in vitro and in vivo We report in vitro PNP-inhibitor dissociation rates (t½) from 3 to 31 min for seven Immucillins with dissociation constants of 115 to 6 pm Treatment of human erythrocytes with DADMe-Immucillin-H (DADMe-ImmH, 22 pm) causes complete inhibition of PNP. Loss of [14C]DADMe-ImmH from erythrocytes during multiple washes is slow and biphasic, resulting from inhibitor release and rebinding to PNP catalytic sites. The slow phase gave a t½ of 84 h. Loss of [14C]DADMe-ImmH from erythrocytes in the presence of excess unlabeled DADMe-ImmH increased to a t½ of 1.6 h by preventing rebinding. Thus, in human erythrocytes, rebinding of DADMe-ImmH is 50-fold more likely than diffusional loss of the inhibitor from the erythrocyte. Humans treated with a single oral dose of DADMe-ImmH in phase 1 clinical trials exhibit regain of PNP activity with a t½ of 59 days, corresponding to the erythropoiesis rate in humans. Thus, the PNP catalytic site recapture of DADMe-ImmH is highly favored in vivo We conclude that transition-state analogues with picomolar dissociation constants exhibit long lifetimes on their targets in vivo because the probability of the target enzyme recapturing inhibitor molecules is greater than diffusional loss to the extracellular space.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Espacio Intracelular/metabolismo , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Transporte Biológico , Ensayos Clínicos Fase I como Asunto , Enzimas , Eritrocitos/efectos de los fármacos , Eritrocitos/enzimología , Humanos , Espacio Intracelular/efectos de los fármacos , Unión Proteica , Purina-Nucleósido Fosforilasa/metabolismo , Pirrolidinas/metabolismo , Pirrolidinas/farmacología
4.
Allergy ; 71(12): 1676-1683, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27154593

RESUMEN

BACKGROUND: Avoralstat is a potent small-molecule oral plasma kallikrein inhibitor under development for treatment of hereditary angioedema (HAE). This first-in-human study evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of avoralstat. METHODS: This double-blind, placebo-controlled, ascending-dose cohort trial evaluated avoralstat single doses of 50, 125, 250, 500, and 1000 mg and multiple doses up to 2400 mg daily (100, 200, 400, and 800 mg every 8 h [q8 h] up to 7 days). RESULTS: Avoralstat (n = 71) was generally well tolerated with no signals for a safety concern; there were no serious adverse events (AEs) or discontinuations due to AEs, and compared to placebo (n = 18), no notable difference in AEs. Four moderate severity AEs were reported in two subjects; syncope after a single 250 mg dose (one subject) and abdominal pain, back pain, and eczema after multiple doses of 800 mg avoralstat (one subject). For multiple-dose cohorts, the incidence of gastrointestinal AEs was highest at the 2400 mg/day dose. Elimination of avoralstat was bi-exponential with a terminal half-life of 12-31 h. Inhibition of plasma kallikrein was observed at all doses, and the degree of inhibition was highly correlated with avoralstat concentrations (R = 0.93). Mean avoralstat concentrations at doses ≥400 mg q8 h met or exceeded plasma kallikrein EC50 values throughout the dosing interval. CONCLUSION: Avoralstat was well tolerated, and drug exposure was sufficient to meet target levels for inhibition of plasma kallikrein. Based on these results, the 400 mg q8 h dose was selected for further evaluation in patients with HAE.


Asunto(s)
Angioedemas Hereditarios/tratamiento farmacológico , Calicreína Plasmática/antagonistas & inhibidores , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacocinética , Administración Oral , Adolescente , Adulto , Esquema de Medicación , Monitoreo de Drogas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Proteasas/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
5.
Eur J Med Chem ; 264: 115991, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38118393

RESUMEN

Hepatitis C infection is caused by the bloodborne pathogen hepatitis C virus (HCV) and can lead to serious liver diseases and, ultimately, death if the treatment is ineffective. This work reports the synthesis and preclinical evaluation of 7 novel 9-O/N/S pyrimidine nucleosides, including compound 12, the triphosphate of known compound 7b. The nucleosides are 9-deaza modifications of adenosine and guanosine with ß-2'-C-methyl substituent on the ribose. Within this series of compounds, a 9-deaza furopyrimidine analog of adenosine, compound 7b, showed high anti-HCV activity in vitro, good stability, low toxicity, and low genotoxicity when administrated in low doses, and an adequate pharmacokinetics profile. An improved synthesis of compound 7b compared to a previous study is also reported. Compound 12 was synthesized as a control to verify phosphorylation of 7b occurred in vivo.


Asunto(s)
Hepatitis C , Nucleósidos de Pirimidina , Humanos , Nucleósidos/farmacología , Hepacivirus , ARN Polimerasa Dependiente del ARN , Nucleósidos de Pirimidina/farmacología , Hepatitis C/tratamiento farmacológico , Adenosina , Antivirales
6.
Antiviral Res ; 195: 105180, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34551346

RESUMEN

Galidesivir (BCX4430) is an adenosine nucleoside analog that is broadly active in cell culture against several RNA viruses of various families. This activity has also been shown in animal models of viral disease associated with Ebola, Marburg, yellow fever, Zika, and Rift Valley fever viruses. In many cases, the compound is more efficacious in animal models than cell culture activity would predict. Based on favorable data from in vivo animal studies, galidesivir has recently undergone evaluation in several phase I clinical trials, including against severe acute respiratory syndrome coronavirus 2, and as a medical countermeasure for the treatment of Marburg virus disease.


Asunto(s)
Adenina/análogos & derivados , Adenosina/análogos & derivados , Antivirales/farmacología , Pirrolidinas/farmacología , Adenina/farmacología , Adenosina/farmacología , Animales , Ensayos Clínicos Fase I como Asunto , Evaluación Preclínica de Medicamentos , Marburgvirus/efectos de los fármacos , Nucleósidos/análogos & derivados , SARS-CoV-2/efectos de los fármacos
7.
Antiviral Res ; 137: 14-22, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27838352

RESUMEN

Zika virus (ZIKV) is currently undergoing pandemic emergence. While disease is typically subclinical, severe neurologic manifestations in fetuses and newborns after congenital infection underscore an urgent need for antiviral interventions. The adenosine analog BCX4430 has broad-spectrum activity against a wide range of RNA viruses, including potent in vivo activity against yellow fever, Marburg and Ebola viruses. We tested this compound against African and Asian lineage ZIKV in cytopathic effect inhibition and virus yield reduction assays in various cell lines. To further evaluate the efficacy in a relevant animal model, we developed a mouse model of severe ZIKV infection, which recapitulates various human disease manifestations including peripheral virus replication, conjunctivitis, encephalitis and myelitis. Time-course quantification of viral RNA accumulation demonstrated robust viral replication in several relevant tissues, including high and persistent viral loads observed in the brain and testis. The presence of viral RNA in various tissues was confirmed by an infectious culture assay as well as immunohistochemical staining of tissue sections. Treatment of ZIKV-infected mice with BCX4430 significantly improved outcome even when treatment was initiated during the peak of viremia. The demonstration of potent activity of BCX4430 against ZIKV in a lethal mouse model warrant its continued clinical development.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Nucleósidos de Purina/farmacología , Nucleósidos de Purina/uso terapéutico , Infección por el Virus Zika/tratamiento farmacológico , Virus Zika/efectos de los fármacos , Adenina/análogos & derivados , Adenosina/análogos & derivados , Animales , Antivirales/administración & dosificación , Encéfalo/virología , Línea Celular , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Nucleósidos de Purina/administración & dosificación , Pirrolidinas , ARN Viral , Testículo/virología , Carga Viral/efectos de los fármacos , Viremia , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/virología
8.
Med Chem ; 2(6): 545-53, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17105435

RESUMEN

Uncontrolled kallikrein activation is involved in diseases such as hereditary angioedema, bacterial septic shock and procedures such as cardiopulmonary bypass. Here we report a series of small molecule compounds that potently inhibit kallikrein activity in vitro. Kinetic studies indicate that some of these compounds are slow binding inhibitors of kallikrein with Ki final less than a nanomolar. The ability of these compounds to inhibit the activity of kallikrein was further confirmed in a plasma model by quantitating the release of bradykinin, an endogenous cleavage product of plasma kallikrein. To understand the inhibitory mechanism of the selected compounds toward kallikrein, the interactions between the selected compounds and kallikrein was explored using molecular modeling based on the information of crystal structures of TF/FVIIa and kallikrein. The information presented in the current study provides an initial approach to develop more selective and therapeutically useful small molecule inhibitors.


Asunto(s)
Calicreínas/antagonistas & inhibidores , Bradiquinina/análisis , Dominio Catalítico , Factor VIIa , Humanos , Calicreínas/química , Cinética , Modelos Moleculares , Plasma/metabolismo , Unión Proteica , Tromboplastina
10.
J Mol Biol ; 204(1): 191-204, 1988 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-3145979

RESUMEN

The crystal structure of mammalian calmodulin has been refined at 2.2 A (1 A = 0.1 nm) resolution using a restrained least-squares method. The final crystallographic R-factor, based on 6685 reflections in the range 2.2 A less than or equal to d less than or equal to 5.0 A with intensities exceeding 2.5 sigma, is 0.175. Bond lengths and bond angles in the molecule have root-mean-square deviations from ideal values of 0.016 A and 1.7 degrees, respectively. The refined model includes residues 5 to 147, four Ca2+ and 69 water molecules per molecule of calmodulin. The electron density for residues 1 to 4 and 148 is poorly defined, and they are not included in the model. The molecule is shaped somewhat like a dumbbell, with an overall length of 65 A; the two lobes are connected by a seven-turn alpha-helix. Prominent secondary structural features include seven alpha-helices, four Ca2+-binding loops, and two short, double-stranded antiparallel beta-sheets between pairs of adjacent Ca2+-binding loops. The four Ca2+-binding domains in calmodulin have a typical EF hand conformation (helix-loop-helix) and are similar to those described in other Ca2+-binding proteins. The X-ray structure determination of calmodulin shows a large hydrophobic cleft in each half of the molecule. These hydrophobic regions probably represent the sites of interaction with many of the pharmacological agents known to bind to calmodulin.


Asunto(s)
Calmodulina , Secuencia de Aminoácidos , Animales , Calcio , Bovinos , Lisina , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Agua , Difracción de Rayos X
11.
J Mol Biol ; 221(4): 1071-3, 1991 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-1942041

RESUMEN

Crystals of a sarcoplasmic Ca(2+)-binding protein from the protochordate amphioxus have been grown from solutions of ammonium sulfate. The crystals are orthorhombic, space group C222(1), with unit cell axes a = 59.6(1) A, b = 81.3(1) A and c = 82.4(1) A. There is one molecule in the asymmetric unit. The crystals diffract beyond 2.5 A and show less than 20% decline in diffraction intensities after a three day exposure to X-rays from a laboratory rotating anode source.


Asunto(s)
Proteínas de Unión al Calcio/química , Cordados no Vertebrados/química , Retículo Sarcoplasmático/química , Sulfato de Amonio/química , Animales , Cristalización , Cristalografía , Difracción de Rayos X
12.
J Mol Biol ; 232(4): 1069-83, 1993 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-8371267

RESUMEN

We present here the three-dimensional structure of neuraminidase (E.C. 3.2.1.18) from influenza virus A/Tern/Australia/G70c/75 (N9), determined by the method of multiple isomorphous replacement, and the structure of the neuraminidase complexed with an inhibitor, 2-deoxy-2,3-dehydro-N-acetyl neuraminic acid (DANA). Native and inhibitor complex crystals are isomorphous and belong to space group I432 with unit cell dimensions of 183.78 A. The native enzyme structure and the inhibitor complex structure have been refined at 2.5 A and 2.8 A resolution, respectively, with crystallographic R-factor values of 0.193 for the native enzyme, and 0.179 for the inhibitor complex. The current enzyme model includes 387 amino acid residues which comprise the asymmetric unit. The root-mean-square deviation from ideal values is 0.013 A for bond lengths and 1.6 degree for bond angles. The neuraminidase (NA), as proteolytically cleaved from the virus, retains full enzymatic and antigenic activity, and is a box-shaped tetramer with edge lengths of 90 A and a maximal depth of 60 A. The NA tetramers are composed of crystallographically equivalent monomers related by circular 4-fold symmetry. Each monomer folds into six antiparallel beta-sheets of four strands. The secondary structure composition is 50% beta-sheet. The remaining 50% of the residues form 24 strand-connecting loops or turns. One of the loops contains a small alpha-helix. The structure of the complex of NA with DANA, a transition state analog, has enabled us to identify and characterize the site of enzyme catalysis. The center of mass of bound inhibitor is 32 A from the 4-fold axis of the tetramer, lodged at the end of a shallow crater of diameter 16 A with a depth of 8 to 10 A. There are 12 amino acid residues that directly bind DANA, with a further six conserved amino acids lining the active site pocket. The neuraminidase inhibitor complex provides a three-dimensional model which will be used to further the understanding of enzymatic hydrolysis and aid the design of specific, antineuraminidase antiviral compounds.


Asunto(s)
Virus de la Influenza A/enzimología , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/química , Ácidos Siálicos/química , Sitios de Unión , Virus de la Influenza B/enzimología , Mercurio/química , Modelos Moleculares , Conformación Molecular , Ácido N-Acetilneuramínico , Platino (Metal)/química , Conformación Proteica , Ácidos Siálicos/metabolismo , Difracción de Rayos X
13.
J Mol Biol ; 196(3): 741-2, 1987 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-3681976

RESUMEN

Two different crystal forms of human C-reactive protein have been grown from solutions of 2-methyl-2,4-pentanediol. Both crystal forms are tetragonal, the space group for form I is P4(1)22 (or P4(3)22), and that for form II is P4(2)22. The unit cell parameters for form I are a = b = 103.0(5) A, c = 308.5(7) A and for form II are a = b = 103.1(2) A, c = 312.7(6) A. The crystals of form II diffract to at least 3.0 A resolution, and are suitable for detailed structural studies.


Asunto(s)
Proteína C-Reactiva , Cristalografía , Humanos
14.
J Mol Biol ; 216(3): 491-6, 1990 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-2258923

RESUMEN

Rotation function studies of two tetragonal crystal forms of human C-reactive protein have confirmed the pentameric structure of the molecule. The two crystal forms have space groups P4122 (I) and P4222 (II) with closely similar unit cells and are often twinned together. Investigation of the crystallization conditions indicates that dissociation heterogeneity has been a major limiting factor in the reproducible growth of good single crystals. The orientation of the pentameric molecule is shown to be almost identical in both forms, about the axial direction omega = 57 degrees, phi = 45 degrees, i.e. 57 degrees away from c in the (110) plane.


Asunto(s)
Proteína C-Reactiva/química , Humanos , Conformación Proteica , Difracción de Rayos X
15.
J Mol Biol ; 219(1): 1-3, 1991 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-2023254

RESUMEN

Human factor D, an essential enzyme of the alternative pathway of complement activation, has been crystallized. Crystals were grown by vapor diffusion using polyethylene glycol 6000 and NaCl as precipitants. The factor D crystals are triclinic and the space group is P1 with unit cell dimensions a = 40.8 A, b = 64.7 A, c = 40.3 A, alpha = 101.0 degrees, beta = 109.7 degrees, gamma = 74.3 degrees. The unit cell contains two molecules of factor D related by a non-crystallographic 2-fold axis. The crystals grow to dimensions of 0.8 mm x 0.5 mm x 0.2 mm within five days, are stable in the X-ray beam and diffract beyond 2.5 A.


Asunto(s)
Factor D del Complemento/química , Factor D del Complemento/aislamiento & purificación , Cristalización , Síndrome de Fanconi/orina , Humanos , Conformación Proteica , Difracción de Rayos X/métodos
16.
J Mol Biol ; 282(5): 1061-81, 1998 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-9753554

RESUMEN

Factor D is a serine protease essential for the activation of the alternative pathway of complement. The structures of native factor D and a complex formed with isatoic anhydride inhibitor were determined at resolution of 2.3 and 1.5 A, respectively, in an isomorphous monoclinic crystal form containing one molecule per asymmetric unit. The native structure was compared with structures determined previously in a triclinic cell containing two molecules with different active site conformations. The current structure shows greater similarity with molecule B in the triclinic cell, suggesting that this may be the dominant factor D conformation in solution. The major conformational differences with molecule A in the triclinic cell are located in four regions, three of which are close to the active site and include some of the residues shown to be critical for factor D catalytic activity. The conformational flexibility associated with these regions is proposed to provide a structural basis for the previously proposed substrate-induced reversible conformational changes in factor D. The high-resolution structure of the factor D/isatoic anhydride complex reveals the binding mode of the mechanism-based inhibitor. The higher specificity towards factor D over trypsin and thrombin is based on hydrophobic interactions between the inhibitor benzyl ring and the aliphatic side-chain of Arg218 that is salt bridged with Asp189 at the bottom of the primary specificity (S1) pocket. Comparison of factor D structural variants with other serine protease structures revealed the presence of a unique "self-inhibitory loop". This loop (214-218) dictates the resting-state conformation of factor D by (1) preventing His57 from adopting active tautomer conformation, (2) preventing the P1 to P3 residues of the substrate from forming anti-parallel beta-sheets with the non-specific substrate binding loop, and (3) blocking the accessibility of Asp189 to the positive1y charged P1 residue of the substrate. The conformational switch from resting-state to active-state can only be induced by the single macromolecular substrate, C3b-bound factor B. This self-inhibitory mechanism is highly correlated with the unique functional properties of factor D, which include high specificity toward factor B, low esterolytic activity toward synthetic substrates, and absence of regulation by zymogen and serpin-like or other natural inhibitors in blood.


Asunto(s)
Factor D del Complemento/química , Factor D del Complemento/metabolismo , Oxazinas/química , Sitios de Unión , Cumarinas/química , Cumarinas/metabolismo , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Histidina , Isocumarinas , Modelos Moleculares , Oxazinas/metabolismo , Conformación Proteica , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/metabolismo , Agua
17.
J Mol Biol ; 170(2): 529-65, 1983 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-6415289

RESUMEN

The binding sites for the allosteric activator, AMP, to glycogen phosphorylase b are described in detail utilizing the more precise knowledge of the native structure obtained from crystallographic restrained least-squares refinement than has hitherto been available. Localized conformational changes are seen at the allosteric effector site that include shifts of between 1 and 2 A for residues Tyr75 and Arg309 and very small shifts for the region of residues 42 to 44 from the symmetry-related subunit. Kinetic studies demonstrate that NADH inhibits the AMP activation of glycogen phosphorylase b. Crystallographic binding studies at 3.5 A resolution show that NADH binds to the same sites on the enzyme as AMP, i.e. the allosteric effector site N, which is close to the subunit-subunit interface, and the nucleoside inhibitor site I, which is some 12 A from the catalytic site. The conformations of NADH at the two sites are different but both conformations are "folded" so that the nicotinamide ring is close (approx. 6 A) to the adenine ring. These conformations are compared with those suggested from solution studies and with the extended conformations observed in the single crystal structure of NAD+ and for NAD bound to dehydrogenases. Possible mechanisms for NADH inhibition of phosphorylase activation are discussed.


Asunto(s)
Adenosina Monofosfato/metabolismo , NAD/metabolismo , Fosforilasa b/metabolismo , Fosforilasas/metabolismo , Sitio Alostérico , Animales , Sitios de Unión , Cristalografía , Activación Enzimática/efectos de los fármacos , Cinética , Sustancias Macromoleculares , Conformación Molecular , Conformación Proteica , Conejos
18.
J Mol Biol ; 218(4): 695-8, 1991 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-1902521

RESUMEN

Large crystals of porcine aldose reductase have been grown from polyethylene glycol solutions. The crystals are triclinic, space-group P1, with a = 81.3 A, b = 85.9 A, c = 56.6 A, alpha = 102.3 degrees, beta = 103.3 degrees and gamma = 79.0 degrees. The crystals grow within ten days to dimensions of 0.6 mm x 0.4 mm x 0.2 mm and diffract to at least 2.5 A. There are four molecules in the unit cell related by a set of three mutually perpendicular non-crystallographic 2-fold axes.


Asunto(s)
Aldehído Reductasa/química , Aldehído Reductasa/aislamiento & purificación , Animales , Conformación Proteica , Porcinos , Difracción de Rayos X
19.
J Mol Biol ; 267(3): 584-94, 1997 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-9126840

RESUMEN

The active site of influenza virus neuraminidase (NA) is formed by 11 universally conserved residues. A guanidino group incorporated into two unrelated NA inhibitors was previously reported to occupy different negatively charged sites in the NA active site, A new inhibitor containing two guanidino groups was synthesized in order to utilize both sites in an attempt to acquire a combined increase in affinity. The X-ray crystal structures of the complexes show that the expected increase in affinity could not be achieved even though the added guanidino group binds to the negatively charged site as designed. This suggests that the ligand affinity to the target protein is contributed both from ligand-protein interactions and solvation/conformation energy of the ligand.


Asunto(s)
Guanidinas/farmacología , Hidroxibenzoatos/farmacología , Virus de la Influenza B/enzimología , Neuraminidasa/química , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Guanidinas/química , Humanos , Hidroxibenzoatos/química , Modelos Moleculares , Neuraminidasa/antagonistas & inhibidores , Agua/química
20.
J Med Chem ; 36(24): 3771-83, 1993 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-8254607

RESUMEN

X-ray crystallography and computer-assisted molecular modeling (CAMM) studies aided in the design of a potent series of mammalian purine nucleoside phosphorylase (PNP) inhibitors. Enhanced potency was achieved by designing substituted 9-(arylmethyl)-9-deazaguanine analogs that interact favorably with all three of the binding subsites of the PNP active site, namely the purine binding site, the hydrophobic pocket, and the phosphate binding site. The most potent PNP inhibitor prepared during our investigation, (S)-9-[1-(3-chlorophenyl)-2-carboxyethyl]-9-deazaguanine (18b), was shown to have an IC50 of 6 nM, whereas the corresponding (R)-isomer was 30-fold less potent.


Asunto(s)
Guanina/análogos & derivados , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Pirimidinas/síntesis química , Pirroles/síntesis química , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Guanina/química , Guanina/farmacología , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Fosfatos/metabolismo , Purinas/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA