Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Biochemistry ; 56(3): 458-467, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28029774

RESUMEN

N1-Acetylspermine oxidase (APAO) catalyzes the conversion of N1-acetylspermine or N1-acetylspermidine to spermidine or putrescine, respectively, with concomitant formation of N-acetyl-3-aminopropanal and hydrogen peroxide. Here we present the structure of murine APAO in its oxidized holo form and in complex with substrate. The structures provide a basis for understanding molecular details of substrate interaction in vertebrate APAO, highlighting a key role for an asparagine residue in coordinating the N1-acetyl group of the substrate. We applied computational methods to the crystal structures to rationalize previous observations with regard to the substrate charge state. The analysis suggests that APAO features an active site ideally suited for binding of charged polyamines. We also reveal the structure of APAO in complex with the irreversible inhibitor MDL72527. In addition to the covalent adduct, a second MDL72527 molecule is bound in the active site. Binding of MDL72527 is accompanied by altered conformations in the APAO backbone. On the basis of structures of APAO, we discuss the potential for development of specific inhibitors.


Asunto(s)
Oxidorreductasas/química , Putrescina/química , Espermidina/análogos & derivados , Espermidina/química , Espermina/análogos & derivados , Aldehídos/química , Aldehídos/metabolismo , Animales , Dominio Catalítico , Expresión Génica , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Cinética , Ratones , Modelos Moleculares , Oxidación-Reducción , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Propilaminas/química , Propilaminas/metabolismo , Estructura Secundaria de Proteína , Putrescina/análogos & derivados , Putrescina/metabolismo , Espermidina/metabolismo , Espermina/química , Espermina/metabolismo
2.
Blood ; 125(22): 3484-90, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25788700

RESUMEN

Ticagrelor is a direct-acting reversibly binding P2Y12 antagonist and is widely used as an antiplatelet therapy for the prevention of cardiovascular events in acute coronary syndrome patients. However, antiplatelet therapy can be associated with an increased risk of bleeding. Here, we present data on the identification and the in vitro and in vivo pharmacology of an antigen-binding fragment (Fab) antidote for ticagrelor. The Fab has a 20 pM affinity for ticagrelor, which is 100 times stronger than ticagrelor's affinity for its target, P2Y12. Despite ticagrelor's structural similarities to adenosine, the Fab is highly specific and does not bind to adenosine, adenosine triphosphate, adenosine 5'-diphosphate, or structurally related drugs. The antidote concentration-dependently neutralized the free fraction of ticagrelor and reversed its antiplatelet activity both in vitro in human platelet-rich plasma and in vivo in mice. Lastly, the antidote proved effective in normalizing ticagrelor-dependent bleeding in a mouse model of acute surgery. This specific antidote for ticagrelor may prove valuable as an agent for patients who require emergency procedures.


Asunto(s)
Adenosina/análogos & derivados , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/farmacología , Antídotos/química , Antídotos/farmacología , Adenosina/antagonistas & inhibidores , Adenosina/inmunología , Animales , Anticuerpos/aislamiento & purificación , Anticuerpos/metabolismo , Especificidad de Anticuerpos , Anticuerpos ampliamente neutralizantes , Células CHO , Cricetinae , Cricetulus , Cristalografía por Rayos X , Hemorragia/prevención & control , Humanos , Fragmentos Fab de Inmunoglobulinas/farmacología , Ratones , Modelos Moleculares , Agregación Plaquetaria/efectos de los fármacos , Ingeniería de Proteínas , Ticagrelor
3.
Proc Natl Acad Sci U S A ; 110(10): 3806-11, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23431194

RESUMEN

Prostaglandin E2 (PGE2) is a key mediator in inflammatory response. The main source of inducible PGE2, microsomal PGE2 synthase-1 (mPGES-1), has emerged as an interesting drug target for treatment of pain. To support inhibitor design, we have determined the crystal structure of human mPGES-1 to 1.2 Å resolution. The structure reveals three well-defined active site cavities within the membrane-spanning region in each monomer interface of the trimeric structure. An important determinant of the active site cavity is a small cytosolic domain inserted between transmembrane helices I and II. This extra domain is not observed in other structures of proteins within the MAPEG (Membrane-Associated Proteins involved in Eicosanoid and Glutathione metabolism) superfamily but is likely to be present also in microsomal GST-1 based on sequence similarity. An unexpected feature of the structure is a 16-Å-deep cone-shaped cavity extending from the cytosolic side into the membrane-spanning region. We suggest a potential role for this cavity in substrate access. Based on the structure of the active site, we propose a catalytic mechanism in which serine 127 plays a key role. We have also determined the structure of mPGES-1 in complex with a glutathione-based analog, providing insight into mPGES-1 flexibility and potential for structure-based drug design.


Asunto(s)
Oxidorreductasas Intramoleculares/química , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Glutatión/análogos & derivados , Glutatión/química , Humanos , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/genética , Microsomas/enzimología , Modelos Moleculares , Datos de Secuencia Molecular , Prostaglandina-E Sintasas , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido
4.
J Biol Chem ; 288(2): 873-85, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23155046

RESUMEN

A novel class of small molecule inhibitors for plasminogen activator inhibitor type 1 (PAI-1), represented by AZ3976, was identified in a high throughput screening campaign. AZ3976 displayed an IC(50) value of 26 µm in an enzymatic chromogenic assay. In a plasma clot lysis assay, the compound was active with an IC(50) of 16 µm. Surprisingly, AZ3976 did not bind to active PAI-1 but bound to latent PAI-1 with a K(D) of 0.29 µm at 35 °C and a binding stoichiometry of 0.94, as measured by isothermal calorimetry. Reversible binding was confirmed by surface plasmon resonance direct binding experiments. The x-ray structure of AZ3976 in complex with latent PAI-1 was determined at 2.4 Å resolution. The inhibitor was bound in the flexible joint region with the entrance to the cavity located between α-helix D and ß-strand 2A. A set of surface plasmon resonance experiments revealed that AZ3976 inhibited PAI-1 by enhancing the latency transition of active PAI-1. Because AZ3976 only had measurable affinity for latent PAI-1, we propose that its mechanism of inhibition is based on binding to a small fraction in equilibrium with active PAI-1, a latent-like prelatent form, from which latent PAI-1 is then generated more rapidly. This mode of action, with induced accelerated latency transition of active PAI-1 may, together with supporting x-ray data, provide improved opportunities for small molecule drug design in the hunt for therapeutically useful PAI-1 inhibitors.


Asunto(s)
Azetidinas/farmacología , Inhibidor 1 de Activador Plasminogénico/química , Pirimidinonas/farmacología , Animales , Azetidinas/química , Células CHO , Calorimetría , Cricetinae , Cricetulus , Humanos , Modelos Moleculares , Conformación Proteica , Pirimidinonas/química , Ratas , Resonancia por Plasmón de Superficie , Termodinámica
5.
J Biol Chem ; 288(51): 36636-47, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24194519

RESUMEN

The neutrophil enzyme myeloperoxidase (MPO) promotes oxidative stress in numerous inflammatory pathologies by producing hypohalous acids. Its inadvertent activity is a prime target for pharmacological control. Previously, salicylhydroxamic acid was reported to be a weak reversible inhibitor of MPO. We aimed to identify related hydroxamates that are good inhibitors of the enzyme. We report on three hydroxamates as the first potent reversible inhibitors of MPO. The chlorination activity of purified MPO was inhibited by 50% by a 5 nm concentration of a trifluoromethyl-substituted aromatic hydroxamate, HX1. The hydroxamates were specific for MPO in neutrophils and more potent toward MPO compared with a broad range of redox enzymes and alternative targets. Surface plasmon resonance measurements showed that the strength of binding of hydroxamates to MPO correlated with the degree of enzyme inhibition. The crystal structure of MPO-HX1 revealed that the inhibitor was bound within the active site cavity above the heme and blocked the substrate channel. HX1 was a mixed-type inhibitor of the halogenation activity of MPO with respect to both hydrogen peroxide and halide. Spectral analyses demonstrated that hydroxamates can act variably as substrates for MPO and convert the enzyme to a nitrosyl ferrous intermediate. This property was unrelated to their ability to inhibit MPO. We propose that aromatic hydroxamates bind tightly to the active site of MPO and prevent it from producing hypohalous acids. This mode of reversible inhibition has potential for blocking the activity of MPO and limiting oxidative stress during inflammation.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hidrocarburos Aromáticos/farmacología , Ácidos Hidroxámicos/farmacología , Peroxidasa/química , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Inhibidores Enzimáticos/química , Humanos , Hidrocarburos Aromáticos/síntesis química , Hidrocarburos Aromáticos/química , Ácidos Hidroxámicos/química , Cinética , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Neutrófilos/enzimología , Peroxidasa/antagonistas & inhibidores , Peroxidasa/metabolismo , Unión Proteica
6.
J Biol Chem ; 286(43): 37578-89, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21880720

RESUMEN

Myeloperoxidase (MPO) is a prime candidate for promoting oxidative stress during inflammation. This abundant enzyme of neutrophils uses hydrogen peroxide to oxidize chloride to highly reactive and toxic chlorine bleach. We have identified 2-thioxanthines as potent mechanism-based inactivators of MPO. Mass spectrometry and x-ray crystal structures revealed that these inhibitors become covalently attached to the heme prosthetic groups of the enzyme. We propose a mechanism whereby 2-thioxanthines are oxidized, and their incipient free radicals react with the heme groups of the enzyme before they can exit the active site. 2-Thioxanthines inhibited MPO in plasma and decreased protein chlorination in a mouse model of peritonitis. They slowed but did not prevent neutrophils from killing bacteria and were poor inhibitors of thyroid peroxidase. Our study shows that MPO is susceptible to the free radicals it generates, and this Achilles' heel of the enzyme can be exploited to block oxidative stress during inflammation.


Asunto(s)
Inhibidores Enzimáticos , Neutrófilos/enzimología , Estrés Oxidativo/efectos de los fármacos , Peritonitis/enzimología , Peroxidasa , Xantinas , Animales , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etnología , Inflamación/microbiología , Inflamación/patología , Ratones , Neutrófilos/patología , Oxidación-Reducción/efectos de los fármacos , Peritonitis/tratamiento farmacológico , Peritonitis/patología , Peroxidasa/antagonistas & inhibidores , Peroxidasa/química , Peroxidasa/metabolismo , Xantinas/química , Xantinas/farmacología
7.
J Med Chem ; 65(17): 11485-11496, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36005476

RESUMEN

Myeloperoxidase is a promising therapeutic target for treatment of patients suffering from heart failure with preserved ejection fraction (HFpEF). We aimed to discover a covalent myeloperoxidase inhibitor with high selectivity for myeloperoxidase over thyroid peroxidase, limited penetration of the blood-brain barrier, and pharmacokinetics suitable for once-daily oral administration at low dose. Structure-activity relationship, biophysical, and structural studies led to prioritization of four compounds for in-depth safety and pharmacokinetic studies in animal models. One compound (AZD4831) progressed to clinical studies on grounds of high potency (IC50, 1.5 nM in vitro) and selectivity (>450-fold vs thyroid peroxidase in vitro), the mechanism of irreversible inhibition, and the safety profile. Following phase 1 studies in healthy volunteers and a phase 2a study in patients with HFpEF, a phase 2b/3 efficacy study of AZD4831 in patients with HFpEF started in 2021.


Asunto(s)
Insuficiencia Cardíaca , Animales , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Yoduro Peroxidasa/uso terapéutico , Peroxidasa , Pirimidinas , Pirroles , Volumen Sistólico/fisiología
8.
Acta Crystallogr D Struct Biol ; 76(Pt 8): 771-777, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32744259

RESUMEN

Advances in synchrotron storage rings and beamline automation have pushed data-collection rates to thousands of data sets per week. With this increase in throughput, massive projects such as in-crystal fragment screening have become accessible to a larger number of research groups. The quality of support offered at large-scale facilities allows medicinal chemistry-focused or biochemistry-focused groups to supplement their research with structural biology. Preparing the experiment, analysing multiple data sets and prospecting for interesting complexes of protein and fragments require, for both newcomers and experienced users, efficient management of the project and extensive computational power for data processing and structure refinement. Here, FragMAX, a new complete platform for fragment screening at the BioMAX beamline of the MAX IV Laboratory, is described. The ways in which users are assisted in X-ray-based fragment screenings and in which the fourth-generation storage ring available at the facility is best exploited are also described.


Asunto(s)
Elementos Estructurales de las Proteínas , Proteínas/química , Programas Informáticos , Automatización , Cristalografía por Rayos X , Recolección de Datos
10.
MAbs ; 7(1): 152-66, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25484051

RESUMEN

Therapeutic monoclonal antibodies targeting G-protein-coupled receptors (GPCRs) are desirable for intervention in a wide range of disease processes. The discovery of such antibodies is challenging due to a lack of stability of many GPCRs as purified proteins. We describe here the generation of Fpro0165, a human anti-formyl peptide receptor 1 (FPR1) antibody generated by variable domain engineering of an antibody derived by immunization of transgenic mice expressing human variable region genes. Antibody isolation and subsequent engineering of affinity, potency and species cross-reactivity using phage display were achieved using FPR1 expressed on HEK cells for immunization and selection, along with calcium release cellular assays for antibody screening. Fpro0165 shows full neutralization of formyl peptide-mediated activation of primary human neutrophils. A crystal structure of the Fpro0165 Fab shows a long, protruding VH CDR3 of 24 amino acids and in silico docking with a homology model of FPR1 suggests that this long VH CDR3 is critical to the predicted binding mode of the antibody. Antibody mutation studies identify the apex of the long VH CDR3 as key to mediating the species cross-reactivity profile of the antibody. This study illustrates an approach for antibody discovery and affinity engineering to typically intractable membrane proteins such as GPCRs.


Asunto(s)
Anticuerpos Monoclonales/química , Regiones Determinantes de Complementariedad/química , Cadenas Pesadas de Inmunoglobulina/química , Simulación del Acoplamiento Molecular , Receptores de Formil Péptido/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Células CHO , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/inmunología , Cricetinae , Cricetulus , Cristalografía por Rayos X , Células HEK293 , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Ratones , Neutrófilos/inmunología , Estructura Cuaternaria de Proteína , Receptores de Formil Péptido/inmunología
11.
Drug Discov Today ; 19(7): 905-11, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24642031

RESUMEN

Cytochrome P450 (CYP) enzymes are key players in xenobiotic metabolism, and inhibition of CYPs can therefore result in unwanted drug-drug interactions. Within drug discovery, CYP inhibition can cause delays in the progression of candidate drugs, or even premature closure of projects. During the past decade, a massive effort in the pharmaceutical industry and academic research has produced a wealth of structural information in the CYP field. In this short review, we will describe how structure-based approaches can be used in the pharmaceutical industry to work away from CYP inhibition, with a focus on the opportunities and challenges. We will show two examples from our own work where structural information on CYP2C9 and CYP3A4 inhibitor complexes have been successfully exploited in ongoing drug discovery projects.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/química , Descubrimiento de Drogas/métodos , Animales , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Ligandos , Estructura Secundaria de Proteína , Relación Estructura-Actividad
12.
J Med Chem ; 57(3): 970-86, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24422550

RESUMEN

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) has been widely considered by the pharmaceutical industry as a target to treat metabolic syndrome in type II diabetics. We hypothesized that central nervous system (CNS) penetration might be required to see efficacy. Starting from a previously reported pyrimidine compound, we removed hydrogen-bond donors to yield 3, which had modest CNS penetration. More significant progress was achieved by changing the core to give 40, which combines good potency and CNS penetration. Compound 40 was dosed to diet-induced obese (DIO) mice and gave excellent target engagement in the liver and high free exposures of drug, both peripherally and in the CNS. However, no body weight reduction or effects on glucose or insulin were observed in this model. Similar data were obtained with a structurally diverse thiazole compound 51. This work casts doubt on the hypothesis that localized tissue modulation of 11ß-HSD1 activity alleviates metabolic syndrome.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Adamantano/análogos & derivados , Adamantano/síntesis química , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/síntesis química , Síndrome Metabólico/tratamiento farmacológico , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , Adamantano/farmacocinética , Adamantano/farmacología , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Cristalografía por Rayos X , Ciclopropanos/síntesis química , Ciclopropanos/farmacocinética , Ciclopropanos/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Grasas de la Dieta/administración & dosificación , Humanos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Insulina/sangre , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Síndrome Metabólico/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Modelos Moleculares , Pirazoles/síntesis química , Pirazoles/farmacocinética , Pirazoles/farmacología , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/farmacocinética , Tiazoles/farmacología
13.
ACS Med Chem Lett ; 3(9): 705-9, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-24900538

RESUMEN

Trk receptor tyrosine kinases have been implicated in cancer and pain. A crystal structure of TrkA with AZ-23 (1a) was obtained, and scaffold hopping resulted in two 5/6-bicyclic series comprising either imidazo[4,5-b]pyridines or purines. Further optimization of these two fusion series led to compounds with subnanomolar potencies against TrkA kinase in cellular assays. Antitumor effects in a TrkA-driven mouse allograft model were demonstrated with compounds 2d and 3a.

15.
J Biol Chem ; 284(16): 10672-83, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19211551

RESUMEN

Cyclotides are a family of plant defense proteins that are highly resistant to adverse chemical, thermal, and enzymatic treatment. Here, we present the first crystal structure of a cyclotide, varv F, from the European field pansy, Viola arvensis, determined at a resolution of 1.8 A. The solution state NMR structure was also determined and, combined with measurements of biophysical parameters for several cyclotides, provided an insight into the structural features that account for the remarkable stability of the cyclotide family. The x-ray data confirm the cystine knot topology and the circular backbone, and delineate a conserved network of hydrogen bonds that contribute to the stability of the cyclotide fold. The structural role of a highly conserved Glu residue that has been shown to regulate cyclotide function was also determined, verifying its involvement in a stabilizing hydrogen bond network. We also demonstrate that varv F binds to dodecylphosphocholine micelles, defining the binding orientation and showing that its structure remains unchanged upon binding, further demonstrating that the cyclotide fold is rigid. This study provides a biological insight into the mechanism by which cyclotides maintain their native activity in the unfavorable environment of predator insect guts. It also provides a structural basis for explaining how a cluster of residues important for bioactivity may be involved in self-association interactions in membranes. As well as being important for their bioactivity, the structural rigidity of cyclotides makes them very suitable as a stable template for peptide-based drug design.


Asunto(s)
Ciclotidas , Motivos Nodales de Cisteina , Diseño de Fármacos , Insecticidas , Conformación Proteica , Pliegue de Proteína , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Ciclotidas/química , Ciclotidas/genética , Ciclotidas/metabolismo , Humanos , Enlace de Hidrógeno , Insecticidas/química , Insecticidas/metabolismo , Datos de Secuencia Molecular , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Alineación de Secuencia
16.
Proc Natl Acad Sci U S A ; 103(37): 13682-7, 2006 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-16954191

RESUMEN

Cytochrome P450 (CYP) 3A4 is the most promiscuous of the human CYP enzymes and contributes to the metabolism of approximately 50% of marketed drugs. It is also the isoform most often involved in unwanted drug-drug interactions. A better understanding of the molecular mechanisms governing CYP3A4-ligand interaction therefore would be of great importance to any drug discovery effort. Here, we present crystal structures of human CYP3A4 in complex with two well characterized drugs: ketoconazole and erythromycin. In contrast to previous reports, the protein undergoes dramatic conformational changes upon ligand binding with an increase in the active site volume by >80%. The structures represent two distinct open conformations of CYP3A4 because ketoconazole and erythromycin induce different types of coordinate shifts. The binding of two molecules of ketoconazole to the CYP3A4 active site and the clear indication of multiple binding modes for erythromycin has implications for the interpretation of the atypical kinetic data often displayed by CYP3A4. The extreme flexibility revealed by the present structures also challenges any attempt to apply computational design tools without the support of relevant experimental data.


Asunto(s)
Sistema Enzimático del Citocromo P-450/química , Eritromicina/química , Cetoconazol/química , Cristalografía , Citocromo P-450 CYP3A , Humanos , Ligandos , Conformación Proteica
17.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 5): 923-5, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15103140

RESUMEN

NtcA is a transcription factor that acts as a global nitrogen regulator in cyanobacteria. Cyanobacteria are photosynthetic prokaryotic organisms, some genera of which can fix nitrogen under conditions of nitrogen deprivation. NtcA from Anabaena PCC 7120 is a dimeric protein that consists of 223 amino acids with a molecular weight of 25 kDa per subunit. It belongs to the cAMP receptor-protein (CAP) family and is involved in the regulation of several of the genes acting in the nitrogen-fixation process. Here, the crystallization and preliminary X-ray data of NtcA are described. The crystallization was made possible by an improved purification method, which provides a stable NtcA protein at concentrations suitable for crystallization. The protein was crystallized using the hanging-drop method. Data were collected to 2.5 A resolution using synchrotron radiation and the crystals belonged to space group P4(1)2(1)2 or P4(3)2(1)2, with unit-cell parameters a = 69.23, b = 69.23, c = 162.15 A, alpha = beta = gamma = 90 degrees. The phases necessary to solve the structure of NtcA could not be obtained by molecular replacement based on the CAP structure using various models.


Asunto(s)
Anabaena/química , Factores de Transcripción/química , Factores de Transcripción/aislamiento & purificación , Cristalización , Dimerización , Conformación Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Factores de Transcripción/genética
18.
J Biol Chem ; 278(14): 11773-81, 2003 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-12556530

RESUMEN

The 1.4-A crystal structure of the oxidized state of a Y25S variant of cytochrome cd(1) nitrite reductase from Paracoccus pantotrophus is described. It shows that loss of Tyr(25), a ligand via its hydroxy group to the iron of the d(1) heme in the oxidized (as prepared) wild-type enzyme, does not result in a switch at the c heme of the unusual bishistidinyl coordination to the histidine/methionine coordination seen in other conformations of the enzyme. The Ser(25) side chain is seen in two positions in the d(1) heme pocket with relative occupancies of approximately 7:3, but in neither case is the hydroxy group bound to the iron atom; instead, a sulfate ion from the crystallization solution is bound between the Ser(25) side chain and the heme iron. Unlike the wild-type enzyme, the Y25S mutant is active as a reductase toward nitrite, oxygen, and hydroxylamine without a reductive activation step. It is concluded that Tyr(25) is not essential for catalysis of reduction of any substrate, but that the requirement for activation by reduction of the wild-type enzyme is related to a requirement to drive the dissociation of this residue from the active site. The Y25S protein retains the d(1) heme less well than the wild-type protein, suggesting that the tyrosine residue has a role in stabilizing the binding of this cofactor.


Asunto(s)
Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Nitrito Reductasas/genética , Nitrito Reductasas/metabolismo , Paracoccus/enzimología , Sitios de Unión/fisiología , Citocromos , Complejo IV de Transporte de Electrones/química , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Hemo/metabolismo , Cinética , Ligandos , Mutagénesis/fisiología , Nitrito Reductasas/química , Estructura Terciaria de Proteína , Serina/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA