Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 142(1): 62-72, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-36796019

RESUMEN

Bruton tyrosine kinase (BTK), a nonreceptor tyrosine kinase, is a major therapeutic target for B-cell-driven malignancies. However, approved covalent BTK inhibitors (cBTKis) are associated with treatment limitations because of off-target side effects, suboptimal oral pharmacology, and development of resistance mutations (eg, C481) that prevent inhibitor binding. Here, we describe the preclinical profile of pirtobrutinib, a potent, highly selective, noncovalent (reversible) BTK inhibitor. Pirtobrutinib binds BTK with an extensive network of interactions to BTK and water molecules in the adenosine triphosphate binding region and shows no direct interaction with C481. Consequently, pirtobrutinib inhibits both BTK and BTK C481 substitution mutants in enzymatic and cell-based assays with similar potencies. In differential scanning fluorimetry studies, BTK bound to pirtobrutinib exhibited a higher melting temperature than cBTKi-bound BTK. Pirtobrutinib, but not cBTKis, prevented Y551 phosphorylation in the activation loop. These data suggest that pirtobrutinib uniquely stabilizes BTK in a closed, inactive conformation. Pirtobrutinib inhibits BTK signaling and cell proliferation in multiple B-cell lymphoma cell lines, and significantly inhibits tumor growth in human lymphoma xenografts in vivo. Enzymatic profiling showed that pirtobrutinib was highly selective for BTK in >98% of the human kinome, and in follow-up cellular studies pirtobrutinib retained >100-fold selectivity over other tested kinases. Collectively, these findings suggest that pirtobrutinib represents a novel BTK inhibitor with improved selectivity and unique pharmacologic, biophysical, and structural attributes with the potential to treat B-cell-driven cancers with improved precision and tolerability. Pirtobrutinib is being tested in phase 3 clinical studies for a variety of B-cell malignancies.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa , Linfoma , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Humanos , Animales , Ensayos Antitumor por Modelo de Xenoinjerto , Linfoma/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Línea Celular Tumoral , Ratones Endogámicos NOD , Masculino , Ratones SCID , Conformación Molecular , Ratones
2.
Nucleic Acids Res ; 44(4): 1813-32, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26826708

RESUMEN

Post-transcriptional control of mitochondrial gene expression, including the processing and generation of mature transcripts as well as their degradation, is a key regulatory step in gene expression in human mitochondria. Consequently, identification of the proteins responsible for RNA processing and degradation in this organelle is of great importance. The metallo-ß-lactamase (MBL) is a candidate protein family that includes ribo- and deoxyribonucleases. In this study, we discovered a function for LACTB2, an orphan MBL protein found in mammalian mitochondria. Solving its crystal structure revealed almost perfect alignment of the MBL domain with CPSF73, as well as to other ribonucleases of the MBL superfamily. Recombinant human LACTB2 displayed robust endoribonuclease activity on ssRNA with a preference for cleavage after purine-pyrimidine sequences. Mutational analysis identified an extended RNA-binding site. Knockdown of LACTB2 in cultured cells caused a moderate but significant accumulation of many mitochondrial transcripts, and its overexpression led to the opposite effect. Furthermore, manipulation of LACTB2 expression resulted in cellular morphological deformation and cell death. Together, this study discovered that LACTB2 is an endoribonuclease that is involved in the turnover of mitochondrial RNA, and is essential for mitochondrial function in human cells.


Asunto(s)
Endorribonucleasas/química , Metaloproteínas/química , Mitocondrias/enzimología , Proteínas de Unión al ARN/química , beta-Lactamasas/química , Sitios de Unión , Cristalografía por Rayos X , Endorribonucleasas/genética , Endorribonucleasas/aislamiento & purificación , Humanos , Metaloproteínas/genética , Estructura Terciaria de Proteína , ARN/genética , ARN Mitocondrial , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/aislamiento & purificación , beta-Lactamasas/genética , beta-Lactamasas/aislamiento & purificación
3.
Protein Expr Purif ; 133: 41-49, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28263854

RESUMEN

Recent innovative approaches to stabilize and crystallize GPCRs have resulted in an unprecedented breakthrough in GPCR crystal structures as well as application of the purified receptor protein in biophysical and biochemical ligand binding assays. However, the protein optimization process to enable these technologies is lengthy and requires iterative overexpression, solubilization, purification and functional analysis of tens to hundreds of protein variants. Here, we report a new and versatile method to screen in parallel hundreds of GPCR variants in HEK293 produced virus-like particles (VLPs) for protein yield, stability, functionality and ligand binding. This approach reduces the time and resources during GPCR construct optimization by eliminating lengthy protein solubilization and purification steps and by its adaptability to many binding assay formats (label or label-free detection). We exemplified the robustness of our VLP method by screening 210 GALR3-VLP variants in a radiometric agonist-based binding assay and a subset of 88 variants in a label-free antagonist-based assay. The resulting GALR3 agonist or antagonist stabilizing variants were then further used for recombinant protein expression in transfected insect cells. The final purified protein variants were successfully immobilized on a biosensor chip and used in a surface plasmon resonance binding assay.


Asunto(s)
Expresión Génica , Receptor de Galanina Tipo 3 , Proteínas Recombinantes de Fusión , Virión , Células HEK293 , Humanos , Estabilidad Proteica , Receptor de Galanina Tipo 3/biosíntesis , Receptor de Galanina Tipo 3/química , Receptor de Galanina Tipo 3/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Virión/química , Virión/genética , Virión/metabolismo
4.
Nucleic Acids Res ; 43(22): 11047-60, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26582912

RESUMEN

The human SNM1A and SNM1B/Apollo proteins are members of an extended family of eukaryotic nuclease containing a motif related to the prokaryotic metallo-ß-lactamase (MBL) fold. SNM1A is a key exonuclease during replication-dependent and transcription-coupled interstrand crosslink repair, while SNM1B/Apollo is required for maintaining telomeric overhangs. Here, we report the crystal structures of SNM1A and SNM1B at 2.16 Å. While both proteins contain a typical MBL-ß-CASP domain, a region of positive charge surrounds the active site of SNM1A, which is absent in SNM1B and explains the greater apparent processivity of SNM1A. The structures of both proteins also reveal a putative, wide DNA-binding groove. Extensive mutagenesis of this groove, coupled with detailed biochemical analysis, identified residues that did not impact on SNM1A catalytic activity, but drastically reduced its processivity. Moreover, we identified a key role for this groove for efficient digestion past DNA interstrand crosslinks, facilitating the key DNA repair reaction catalysed by SNM1A. Together, the architecture and dimensions of this groove, coupled to the surrounding region of high positive charge, explain the remarkable ability of SNM1A to accommodate and efficiently digest highly distorted DNA substrates, such as those containing DNA lesions.


Asunto(s)
Enzimas Reparadoras del ADN/química , ADN/metabolismo , Exodesoxirribonucleasas/química , Proteínas Nucleares/química , Dominio Catalítico , Proteínas de Ciclo Celular , Daño del ADN , Enzimas Reparadoras del ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , Humanos , Modelos Moleculares , Proteínas Nucleares/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
5.
Nucleic Acids Res ; 43(10): 5221-35, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25901030

RESUMEN

Bloom's syndrome helicase (BLM) is a member of the RecQ family of DNA helicases, which play key roles in the maintenance of genome integrity in all organism groups. We describe crystal structures of the BLM helicase domain in complex with DNA and with an antibody fragment, as well as SAXS and domain association studies in solution. We show an unexpected nucleotide-dependent interaction of the core helicase domain with the conserved, poorly characterized HRDC domain. The BLM-DNA complex shows an unusual base-flipping mechanism with unique positioning of the DNA duplex relative to the helicase core domains. Comparison with other crystal structures of RecQ helicases permits the definition of structural transitions underlying ATP-driven helicase action, and the identification of a nucleotide-regulated tunnel that may play a role in interactions with complex DNA substrates.


Asunto(s)
RecQ Helicasas/química , Adenosina Difosfato/química , Cristalografía por Rayos X , ADN/química , Modelos Moleculares , Mutación , Conformación Proteica , Estructura Terciaria de Proteína , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Anticuerpos de Dominio Único/química , Zinc/química
6.
J Biol Chem ; 290(22): 13692-709, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25866208

RESUMEN

Ets transcription factors, which share the conserved Ets DNA-binding domain, number nearly 30 members in humans and are particularly involved in developmental processes. Their deregulation following changes in expression, transcriptional activity, or by chromosomal translocation plays a critical role in carcinogenesis. Ets DNA binding, selectivity, and regulation have been extensively studied; however, questions still arise regarding binding specificity outside the core GGA recognition sequence and the mode of action of Ets post-translational modifications. Here, we report the crystal structures of Etv1, Etv4, Etv5, and Fev, alone and in complex with DNA. We identify previously unrecognized features of the protein-DNA interface. Interactions with the DNA backbone account for most of the binding affinity. We describe a highly coordinated network of water molecules acting in base selection upstream of the GGAA core and the structural features that may account for discrimination against methylated cytidine residues. Unexpectedly, all proteins crystallized as disulfide-linked dimers, exhibiting a novel interface (distant to the DNA recognition helix). Homodimers of Etv1, Etv4, and Etv5 could be reduced to monomers, leading to a 40-200-fold increase in DNA binding affinity. Hence, we present the first indication of a redox-dependent regulatory mechanism that may control the activity of this subset of oncogenic Ets transcription factors.


Asunto(s)
Proteínas E1A de Adenovirus/química , Proteínas de Unión al ADN/química , Disulfuros/química , Proteínas Nucleares/química , Proteínas Proto-Oncogénicas c-ets/química , Proteínas Proto-Oncogénicas/química , Factores de Transcripción/química , Secuencia de Aminoácidos , Cromatografía Liquida , ADN/química , Regulación de la Expresión Génica , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Neoplasias/metabolismo , Oxidación-Reducción , Oxígeno/química , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Ionización de Electrospray
7.
Bioorg Med Chem ; 23(17): 5303-10, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26264842

RESUMEN

Soluble guanylate cyclase (sGC) is a haem containing enzyme that regulates cardiovascular homeostasis and multiple mechanisms in the central and peripheral nervous system. Commonly used inhibitors of sGC activity act through oxidation of the haem moiety, however they also bind haemoglobin and this limits their bioavailability for in vivo studies. We have discovered a new class of small molecule inhibitors of sGC and have characterised a compound designated D12 (compound 10) which binds to the catalytic domain of the enzyme with a KD of 11 µM in a SPR assay.


Asunto(s)
Activadores de Enzimas/química , Activadores de Enzimas/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Quinoxalinas/química , Quinoxalinas/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Dominio Catalítico , Guanilato Ciclasa/química , Guanilato Ciclasa/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Guanilil Ciclasa Soluble
8.
Br J Clin Pharmacol ; 77(5): 839-51, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24028545

RESUMEN

AIM: The aim of this study was to investigate relationships between flavin-containing mono-oxygenase 3 (FMO3) genotype and phenotype (conversion of odorous trimethylamine into non-odorous trimethylamine N-oxide) in a large Japanese cohort suffering from trimethylaminuria. METHODS: Urinary excretion of trimethylamine and trimethylamine N-oxide was determined for 102 volunteers with self-reporting symptoms of trimethylaminuria. For each we determined the sequence of the entire coding region, plus 1.3 kb of flanking intronic and 2.5 kb of the upstream region of the FMO3 gene. The affect of upstream variants on transcription was determined with a reporter gene assay. RESULTS: Seventy-eight subjects were diagnosed as suffering from trimethylaminuria, based on urinary excretion of <90% of total TMA as TMA N-oxide. Of these, 13 were classified as severe, 56 as moderate and nine as mild cases, excreting <43%, 48-70% and 73-83% of trimethylamine as trimethylamine N-oxide, respectively. Twenty-seven mutations were identified in FMO3, 15 in the coding region, of which eight abolish or severely impair FMO3 activity (Pro70Leu, Cys197fsX, Thr201Lys, Arg205Cys, Met260Val, Trp388Ter, Gln470Ter and Arg500Ter), and 12 in the upstream region. The mutations segregate into 19 haplotypes, including four different combinations of upstream mutations, each of which reduces transcriptional activity in comparison with the ancestral upstream sequence of FMO3. CONCLUSIONS: Comparisons of genotype and phenotype reveal that severe trimethylaminuria is caused by loss of function mutations in FMO3. For moderate and mild cases the situation is more complex, with most resulting from factors other than FMO3 genotype. Our results have implications for the diagnosis and management of the disorder.


Asunto(s)
Pueblo Asiatico/genética , Errores Innatos del Metabolismo/genética , Metilaminas/orina , Oxigenasas/genética , Adolescente , Adulto , Anciano , Secuencia de Bases , Niño , Preescolar , Femenino , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Fenotipo , Polimorfismo de Nucleótido Simple
9.
Bioorg Med Chem Lett ; 24(4): 1075-9, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24480469

RESUMEN

Soluble Guanylate Cyclase (sGC) is the receptor for the signalling agent nitric oxide (NO) and catalyses the production of the second messenger cyclic guanosine monophosphate (cGMP) from guanosine triphosphate (GTP). The enzyme is an attractive drug target for small molecules that act in the cardiovascular and pulmonary systems, and has also shown to be a potential target in neurological disorders. We have discovered that 5-(indazol-3-yl)-1,2,4-oxadiazoles activate the enzyme in the absence of added NO and shown they bind to the catalytic domain of the enzyme after development of a surface plasmon resonance assay that allows the biophysical detection of intrinsic binding of ligands to the full length sGC and to a construct of the catalytic domain.


Asunto(s)
Guanilato Ciclasa/metabolismo , Oxadiazoles/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Resonancia por Plasmón de Superficie , Biocatálisis , Dominio Catalítico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Guanosina Monofosfato/biosíntesis , Guanilato Ciclasa/antagonistas & inhibidores , Estructura Molecular , Oxadiazoles/química , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Guanilil Ciclasa Soluble , Relación Estructura-Actividad
10.
J Biol Chem ; 287(31): 26254-67, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22692201

RESUMEN

Human SNM1A and SNM1B/Apollo have both been implicated in the repair of DNA interstrand cross-links (ICLs) by cellular studies, and SNM1B is also required for telomere protection. Here, we describe studies on the biochemical characterization of the SNM1A and SNM1B proteins. The results reveal some fundamental differences in the mechanisms of the two proteins. Both SNM1A and SNM1B digest double-stranded and single-stranded DNA with a 5'-to-3' directionality in a reaction that is stimulated by divalent cations, and both nucleases are inhibited by the zinc chelator o-phenanthroline. We find that SNM1A has greater affinity for single-stranded DNA over double-stranded DNA that is not observed with SNM1B. Although both proteins demonstrate a low level of processivity on low molecular weight DNA oligonucleotide substrates, when presented with high molecular weight DNA, SNM1A alone is rendered much more active, being capable of digesting kilobase-long stretches of DNA. Both proteins can digest past ICLs induced by the non-distorting minor groove cross-linking agent SJG-136, albeit with SNM1A showing a greater capacity to achieve this. This is consistent with the proposal that SNM1A and SNM1B might exhibit some redundancy in ICL repair. Together, our work establishes differences in the substrate selectivities of SNM1A and SNM1B that are likely to be relevant to their in vivo roles and which might be exploited in the development of selective inhibitors.


Asunto(s)
Enzimas Reparadoras del ADN/química , Proteínas de Unión al ADN/química , Proteínas Nucleares/química , Proteínas de Ciclo Celular , Quelantes/química , ADN/química , División del ADN , Daño del ADN , Enzimas Reparadoras del ADN/biosíntesis , Enzimas Reparadoras del ADN/aislamiento & purificación , ADN de Cadena Simple/química , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/aislamiento & purificación , Pruebas de Enzimas , Inhibidores Enzimáticos/química , Escherichia coli , Exodesoxirribonucleasas , Fluoresceína/química , Colorantes Fluorescentes/química , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Magnesio/química , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/aislamiento & purificación , Plásmidos/química , Unión Proteica , ARN/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Especificidad por Sustrato
11.
Sci Rep ; 7(1): 7501, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28790404

RESUMEN

The human genome encodes two active Vaccinia-related protein kinases (VRK), VRK1 and VRK2. These proteins have been implicated in a number of cellular processes and linked to a variety of tumors. However, understanding the cellular role of VRKs and establishing their potential use as targets for therapeutic intervention has been limited by the lack of tool compounds that can specifically modulate the activity of these kinases in cells. Here we identified BI-D1870, a dihydropteridine inhibitor of RSK kinases, as a promising starting point for the development of chemical probes targeting the active VRKs. We solved co-crystal structures of both VRK1 and VRK2 bound to BI-D1870 and of VRK1 bound to two broad-spectrum inhibitors. These structures revealed that both VRKs can adopt a P-loop folded conformation, which is stabilized by different mechanisms on each protein. Based on these structures, we suggest modifications to the dihydropteridine scaffold that can be explored to produce potent and specific inhibitors towards VRK1 and VRK2.


Asunto(s)
Antineoplásicos/química , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pteridinas/química , Secuencia de Aminoácidos , Antineoplásicos/farmacología , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Genoma Humano , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Pteridinas/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Virus Vaccinia/genética , Virus Vaccinia/metabolismo
12.
Data Brief ; 12: 603-607, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28540352

RESUMEN

Galanin Receptor 3 (GALR3) is a G-protein-coupled receptor with a widespread distribution in the brain and plays a role in a variety of physiologic processes including cognition/memory, sensory/pain processing, hormone secretion, and feeding behavior. Therefore, GALR3 is considered an attractive CNS drug target (Freimann et al., 2015) [1]. This dataset contains GALR3 point mutants that improve recombinant protein expression and thermal stability of the receptor contained in virus-like particles (VLPs) or obtained by detergent-purification of baculovirus-infected insect cells. The mutations listed can be grouped in those that improve the stability of the agonist-bound and the antagonist-bound form of the receptor. Protein characteristics in terms of protein expression and thermal stability were comparable between GPCR-VLP and GPCR overexpressing Sf9 cultures. The further analysis and detailed results of these mutants as well as their impact on biophysical assay development for drug discovery can be found in "Method for Rapid Optimization of Recombinant GPCR Protein Expression and Stability using Virus-Like Particles" (Ho et al., 2017) [2].

13.
J Med Chem ; 60(8): 3472-3483, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28406621

RESUMEN

A series of potent thienotriazolopyrimidinone-based PDE1 inhibitors was discovered. X-ray crystal structures of example compounds from this series in complex with the catalytic domain of PDE1B and PDE10A were determined, allowing optimization of PDE1B potency and PDE selectivity. Reduction of hERG affinity led to greater than a 3000-fold selectivity for PDE1B over hERG. 6-(4-Methoxybenzyl)-9-((tetrahydro-2H-pyran-4-yl)methyl)-8,9,10,11-tetrahydropyrido[4',3':4,5]thieno[3,2-e][1,2,4]triazolo[1,5-c]pyrimidin-5(6H)-one was identified as an orally bioavailable and brain penetrating PDE1B enzyme inhibitor with potent memory-enhancing effects in a rat model of object recognition memory.


Asunto(s)
Memoria/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Cristalografía por Rayos X , Descubrimiento de Drogas , Inhibidores de Fosfodiesterasa/química
14.
PLoS One ; 8(3): e57644, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23505436

RESUMEN

Soluble guanylate cyclase (sGC) catalyses the synthesis of cyclic GMP in response to nitric oxide. The enzyme is a heterodimer of homologous α and ß subunits, each of which is composed of multiple domains. We present here crystal structures of a heterodimer of the catalytic domains of the α and ß subunits, as well as an inactive homodimer of ß subunits. This first structure of a metazoan, heteromeric cyclase provides several observations. First, the structures resemble known structures of adenylate cyclases and other guanylate cyclases in overall fold and in the arrangement of conserved active-site residues, which are contributed by both subunits at the interface. Second, the subunit interaction surface is promiscuous, allowing both homodimeric and heteromeric association; the preference of the full-length enzyme for heterodimer formation must derive from the combined contribution of other interaction interfaces. Third, the heterodimeric structure is in an inactive conformation, but can be superposed onto an active conformation of adenylate cyclase by a structural transition involving a 26° rigid-body rotation of the α subunit. In the modelled active conformation, most active site residues in the subunit interface are precisely aligned with those of adenylate cyclase. Finally, the modelled active conformation also reveals a cavity related to the active site by pseudo-symmetry. The pseudosymmetric site lacks key active site residues, but may bind allosteric regulators in a manner analogous to the binding of forskolin to adenylate cyclase. This indicates the possibility of developing a new class of small-molecule modulators of guanylate cyclase activity targeting the catalytic domain.


Asunto(s)
Dominio Catalítico , Guanilato Ciclasa/química , Receptores Citoplasmáticos y Nucleares/química , Regulación Alostérica , Aminoácidos/química , Activación Enzimática , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Humanos , Modelos Moleculares , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Subunidades de Proteína , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Recombinantes , Guanilil Ciclasa Soluble
15.
Front Mol Neurosci ; 6: 26, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24068983

RESUMEN

Genetically-encoded biosensors are powerful tools for understanding cellular signal transduction mechanisms. In aiming to investigate cGMP signaling in neurones using the EGFP-based fluorescent biosensor, FlincG (fluorescent indicator for cGMP), we encountered weak or non-existent fluorescence after attempted transfection with plasmid DNA, even in HEK293T cells. Adenoviral infection of HEK293T cells with FlincG, however, had previously proved successful. Both constructs were found to harbor a mutation in the EGFP domain and had a tail of 17 amino acids at the C-terminus that differed from the published sequence. These discrepancies were systematically examined, together with mutations found beneficial for the related GCaMP family of Ca(2+) biosensors, in a HEK293T cell line stably expressing both nitric oxide (NO)-activated guanylyl cyclase and phosphodiesterase-5. Restoring the mutated amino acid improved basal fluorescence whereas additional restoration of the correct C-terminal tail resulted in poor cGMP sensing as assessed by superfusion of either 8-bromo-cGMP or NO. Ultimately, two improved FlincGs were identified: one (FlincG2) had the divergent tail and gave moderate basal fluorescence and cGMP response amplitude and the other (FlincG3) had the correct tail, a GCaMP-like mutation in the EGFP region and an N-terminal tag, and was superior in both respects. All variants tested were strongly influenced by pH over the physiological range, in common with other EGFP-based biosensors. Purified FlincG3 protein exhibited a lower cGMP affinity (0.89 µM) than reported for the original FlincG (0.17 µM) but retained rapid kinetics and a 230-fold selectivity over cAMP. Successful expression of FlincG2 or FlincG3 in differentiated N1E-115 neuroblastoma cells and in primary cultures of hippocampal and dorsal root ganglion cells commends them for real-time imaging of cGMP dynamics in neural (and other) cells, and in their subcellular specializations.

16.
Structure ; 21(7): 1182-92, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23791943

RESUMEN

Malonyl-coenzyme A decarboxylase (MCD) is found from bacteria to humans, has important roles in regulating fatty acid metabolism and food intake, and is an attractive target for drug discovery. We report here four crystal structures of MCD from human, Rhodopseudomonas palustris, Agrobacterium vitis, and Cupriavidus metallidurans at up to 2.3 Å resolution. The MCD monomer contains an N-terminal helical domain involved in oligomerization and a C-terminal catalytic domain. The four structures exhibit substantial differences in the organization of the helical domains and, consequently, the oligomeric states and intersubunit interfaces. Unexpectedly, the MCD catalytic domain is structurally homologous to those of the GCN5-related N-acetyltransferase superfamily, especially the curacin A polyketide synthase catalytic module, with a conserved His-Ser/Thr dyad important for catalysis. Our structures, along with mutagenesis and kinetic studies, provide a molecular basis for understanding pathogenic mutations and catalysis, as well as a template for structure-based drug design.


Asunto(s)
Proteínas Bacterianas/química , Carboxiliasas/química , Mutación Missense , Secuencia de Aminoácidos , Carboxiliasas/deficiencia , Carboxiliasas/genética , Dominio Catalítico , Cristalografía por Rayos X , Enfermedades Carenciales/genética , Estabilidad de Enzimas , Humanos , Enlace de Hidrógeno , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Homología Estructural de Proteína
17.
N Biotechnol ; 29(5): 515-25, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22027370

RESUMEN

The generation of affinity reagents to large numbers of human proteins depends on the ability to express the target proteins as high-quality antigens. The Structural Genomics Consortium (SGC) focuses on the production and structure determination of human proteins. In a 7-year period, the SGC has deposited crystal structures of >800 human protein domains, and has additionally expressed and purified a similar number of protein domains that have not yet been crystallised. The targets include a diversity of protein domains, with an attempt to provide high coverage of protein families. The family approach provides an excellent basis for characterising the selectivity of affinity reagents. We present a summary of the approaches used to generate purified human proteins or protein domains, a test case demonstrating the ability to rapidly generate new proteins, and an optimisation study on the modification of >70 proteins by biotinylation in vivo. These results provide a unique synergy between large-scale structural projects and the recent efforts to produce a wide coverage of affinity reagents to the human proteome.


Asunto(s)
Cromatografía de Afinidad/métodos , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Animales , Biotina/metabolismo , Biotinilación , Cristalización , Medios de Cultivo , Genes , Humanos , Espectrometría de Masas , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Proteoma/genética , Proteoma/aislamiento & purificación , Solubilidad
18.
J Med Chem ; 53(5): 2215-26, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20151671

RESUMEN

We report the molecular design and synthesis of EG00229, 2, the first small molecule ligand for the VEGF-A receptor neuropilin 1 (NRP1) and the structural characterization of NRP1-ligand complexes by NMR spectroscopy and X-ray crystallography. Mutagenesis studies localized VEGF-A binding in the NRP1 b1 domain and a peptide fragment of VEGF-A was shown to bind at the same site by NMR, providing the basis for small molecule design. Compound 2 demonstrated inhibition of VEGF-A binding to NRP1 and attenuated VEGFR2 phosphorylation in endothelial cells. Inhibition of migration of endothelial cells was also observed. The viability of A549 lung carcinoma cells was reduced by 2, and it increased the potency of the cytotoxic agents paclitaxel and 5-fluorouracil when given in combination. These studies provide the basis for design of specific small molecule inhibitors of ligand binding to NRP1.


Asunto(s)
Antineoplásicos/síntesis química , Neuropilina-1/fisiología , Fragmentos de Péptidos/síntesis química , Factor A de Crecimiento Endotelial Vascular/fisiología , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Mutagénesis Sitio-Dirigida , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/ultraestructura , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/ultraestructura , Fosforilación , Relación Estructura-Actividad , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/ultraestructura
19.
Pharmacogenet Genomics ; 17(10): 827-39, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17885620

RESUMEN

OBJECTIVES: Flavin-containing monooxygenase 3 (FMO3) is involved in the metabolism of foreign chemicals, including therapeutic drugs, and thus mediates interactions between humans and their chemical environment. Loss-of-function mutations in the gene cause the inherited disorder trimethylaminuria, or fish-odour syndrome. The objective was to gain insights into the evolutionary history of FMO3. METHODS: Genetic diversity within FMO3 was characterized by sequencing 6.3 kb of genomic DNA, encompassing the entire coding sequence, some intronic and 3'-untranslated region, and 3.4 kb of 5'-flanking sequence, in 23 potential trimethylaminuric Japanese, and the same 3.4 kb 5'-flanking region in 45 unaffected Japanese. Mutational relationships among haplotypes were inferred from a reduced-median network. The time depth of the variation and ages of individual mutations were estimated by maximum-likelihood coalescent analysis. Test statistics were used to investigate whether the variation is compatible with neutral evolution. RESULTS: Sixteen single-nucleotide polymorphisms (SNPs) were identified, which segregated as seven distinct haplotypes. Estimated ages of the mutations indicate that almost all predated migration out of Africa. Analysis of the heterozygosity of FMO3 SNPs indicates that genetic differentiation among continental populations is low (FST=0.050). Test statistics, based on allele-frequency spectrum, number and diversity of haplotypes, linkage disequilibrium and interspecific sequence comparisons, showed a significant departure from neutral expectations, because of an excess of intermediate-frequency SNPs and haplotypes, a ragged pairwise mismatch distribution and an excess of replacement polymorphisms. CONCLUSION: The results provide evidence that FMO3 has been the subject of balancing selection. Finally, we identify mutations that are potential targets for selection.


Asunto(s)
Evolución Molecular , Oxigenasas/genética , Selección Genética , Adulto , Alelos , Secuencia de Bases , Femenino , Frecuencia de los Genes/genética , Haplotipos , Humanos , Desequilibrio de Ligamiento/genética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación/genética , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Recombinación Genética/genética , Análisis de Secuencia de ADN , Especificidad de la Especie , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA