Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Bioorg Med Chem Lett ; 28(9): 1490-1494, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29627262

RESUMEN

Histone lysine demethylases (KDMs) play a key role in epigenetic regulation and KDM5A and KDM5B have been identified as potential anti-cancer drug targets. Using structural information from known KDM4 and KDM5 inhibitors, a potent series of pyrazolylpyridines was designed. Structure-activity relationship (SAR) exploration resulted in the identification of compound 33, an orally available, potent inhibitor of KDM5A/5B with promising selectivity. Potent cellular inhibition as measured by levels of tri-methylated H3K4 was demonstrated with compound 33 in the breast cancer cell line ZR-75-1.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Proteína 2 de Unión a Retinoblastoma/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Femenino , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células MCF-7 , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Modelos Moleculares , Estructura Molecular , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Proteína 2 de Unión a Retinoblastoma/metabolismo , Relación Estructura-Actividad
2.
Bioorg Med Chem Lett ; 28(10): 1811-1816, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29657099

RESUMEN

The bromodomain and extra-terminal (BET) family of epigenetic proteins has attracted considerable attention in drug discovery given its involvement in regulating gene transcription. Screening a focused small molecule library based on the bromodomain pharmacophore resulted in the identification of 2-methylisoquinoline-1-one as a novel BET bromodomain-binding motif. Structure guided SAR exploration resulted in >10,000-fold potency improvement for the BRD4-BD1 bromodomain. Lead compounds exhibited excellent potencies in both biochemical and cellular assays in MYC-dependent cell lines. Compound 36 demonstrated good physicochemical properties and promising exposure levels in exploratory PK studies.


Asunto(s)
Diseño de Fármacos , Isoquinolinas/química , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Sitios de Unión , Proteínas de Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Concentración 50 Inhibidora , Isoquinolinas/síntesis química , Isoquinolinas/farmacología , Simulación de Dinámica Molecular , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
3.
J Biol Chem ; 288(12): 8445-8455, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23355472

RESUMEN

Non-coding apurinic/apyrimidinic (AP) sites in DNA form spontaneously and as DNA base excision repair intermediates are the most common toxic and mutagenic in vivo DNA lesion. For repair, AP sites must be processed by 5' AP endonucleases in initial stages of base repair. Human APE1 and bacterial Nfo represent the two conserved 5' AP endonuclease families in the biosphere; they both recognize AP sites and incise the phosphodiester backbone 5' to the lesion, yet they lack similar structures and metal ion requirements. Here, we determined and analyzed crystal structures of a 2.4 Å resolution APE1-DNA product complex with Mg(2+) and a 0.92 Å Nfo with three metal ions. Structural and biochemical comparisons of these two evolutionarily distinct enzymes characterize key APE1 catalytic residues that are potentially functionally similar to Nfo active site components, as further tested and supported by computational analyses. We observe a magnesium-water cluster in the APE1 active site, with only Glu-96 forming the direct protein coordination to the Mg(2+). Despite differences in structure and metal requirements of APE1 and Nfo, comparison of their active site structures surprisingly reveals strong geometric conservation of the catalytic reaction, with APE1 catalytic side chains positioned analogously to Nfo metal positions, suggesting surprising functional equivalence between Nfo metal ions and APE1 residues. The finding that APE1 residues are positioned to substitute for Nfo metal ions is supported by the impact of mutations on activity. Collectively, the results illuminate the activities of residues, metal ions, and active site features for abasic site endonucleases.


Asunto(s)
Proteínas Bacterianas/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , Desoxirribonucleasa IV (Fago T4-Inducido)/química , Thermotoga maritima/enzimología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , ADN/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Escherichia coli , Humanos , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , Unión Proteica , Estructura Secundaria de Proteína , Homología Estructural de Proteína
4.
Bioorg Med Chem Lett ; 23(6): 1779-85, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23414845

RESUMEN

In order to develop potent and selective focal adhesion kinase (FAK) inhibitors, synthetic studies on pyrazolo[4,3-c][2,1]benzothiazines targeted for the FAK allosteric site were carried out. Based on the X-ray structural analysis of the co-crystal of the lead compound, 8-(4-ethylphenyl)-5-methyl-1,5-dihydropyrazolo[4,3-c][2,1]benzothiazine 4,4-dioxide 1 with FAK, we designed and prepared 1,5-dimethyl-1,5-dihydropyrazolo[4,3-c][2,1]benzothiazin derivatives which selectively inhibited kinase activity of FAK without affecting seven other kinases. The optimized compound, N-(4-tert-butylbenzyl)-1,5-dimethyl-1,5-dihydropyrazolo[4,3-c][2,1]benzothiazin-8-amine 4,4-dioxide 30 possessed significant FAK kinase inhibitory activities both in cell-free (IC50=0.64µM) and in cellular assays (IC50=7.1µM). These results clearly demonstrated a potential of FAK allosteric inhibitors as antitumor agents.


Asunto(s)
Antineoplásicos/química , Óxidos S-Cíclicos/química , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Compuestos Heterocíclicos con 3 Anillos/química , Inhibidores de Proteínas Quinasas/química , Tiazinas/química , Sitio Alostérico , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Óxidos S-Cíclicos/síntesis química , Óxidos S-Cíclicos/metabolismo , Evaluación Preclínica de Medicamentos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Compuestos Heterocíclicos con 3 Anillos/síntesis química , Compuestos Heterocíclicos con 3 Anillos/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Relación Estructura-Actividad , Tiazinas/síntesis química , Tiazinas/metabolismo
5.
Elife ; 112022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35575456

RESUMEN

Chemical manipulation of estrogen receptor alpha ligand binding domain structural mobility tunes receptor lifetime and influences breast cancer therapeutic activities. Selective estrogen receptor modulators (SERMs) extend estrogen receptor alpha (ERα) cellular lifetime/accumulation. They are antagonists in the breast but agonists in the uterine epithelium and/or in bone. Selective estrogen receptor degraders/downregulators (SERDs) reduce ERα cellular lifetime/accumulation and are pure antagonists. Activating somatic ESR1 mutations Y537S and D538G enable resistance to first-line endocrine therapies. SERDs have shown significant activities in ESR1 mutant setting while few SERMs have been studied. To understand whether chemical manipulation of ERα cellular lifetime and accumulation influences antagonistic activity, we studied a series of methylpyrollidine lasofoxifene (Laso) derivatives that maintained the drug's antagonistic activities while uniquely tuning ERα cellular accumulation. These molecules were examined alongside a panel of antiestrogens in live cell assays of ERα cellular accumulation, lifetime, SUMOylation, and transcriptional antagonism. High-resolution x-ray crystal structures of WT and Y537S ERα ligand binding domain in complex with the methylated Laso derivatives or representative SERMs and SERDs show that molecules that favor a highly buried helix 12 antagonist conformation achieve the greatest transcriptional suppression activities in breast cancer cells harboring WT/Y537S ESR1. Together these results show that chemical reduction of ERα cellular lifetime is not necessarily the most crucial parameter for transcriptional antagonism in ESR1 mutated breast cancer cells. Importantly, our studies show how small chemical differences within a scaffold series can provide compounds with similar antagonistic activities, but with greatly different effects of the cellular lifetime of the ERα, which is crucial for achieving desired SERM or SERD profiles.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Ligandos , Mutación , Pirrolidinas , Moduladores Selectivos de los Receptores de Estrógeno/química , Tetrahidronaftalenos
6.
Elife ; 72018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30489256

RESUMEN

Acquired resistance to endocrine therapy remains a significant clinical burden for breast cancer patients. Somatic mutations in the ESR1 (estrogen receptor alpha (ERα)) gene ligand-binding domain (LBD) represent a recognized mechanism of acquired resistance. Antiestrogens with improved efficacy versus tamoxifen might overcome the resistant phenotype in ER +breast cancers. Bazedoxifene (BZA) is a potent antiestrogen that is clinically approved for use in hormone replacement therapies. We found that BZA possesses improved inhibitory potency against the Y537S and D538G ERα mutants compared to tamoxifen and has additional inhibitory activity in combination with the CDK4/6 inhibitor palbociclib. In addition, comprehensive biophysical and structural biology studies show BZA's selective estrogen receptor degrading (SERD) properties that override the stabilizing effects of the Y537S and D538G ERα mutations.


Asunto(s)
Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Receptor alfa de Estrógeno/química , Indoles/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/genética , Femenino , Fulvestrant/farmacología , Humanos , Indoles/química , Ligandos , Células MCF-7 , Proteínas Mutantes/metabolismo , Mutación/genética , Piperazinas/farmacología , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Estructura Secundaria de Proteína , Piridinas/farmacología , Clorhidrato de Raloxifeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/química , Relación Estructura-Actividad , Tamoxifeno/farmacología
7.
Clin Cancer Res ; 24(14): 3433-3446, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29636357

RESUMEN

Purpose: Although high glucocorticoid receptor (GR) expression in early-stage estrogen receptor (ER)-negative breast cancer is associated with shortened relapse-free survival (RFS), how associated GR transcriptional activity contributes to aggressive breast cancer behavior is not well understood. Using potent GR antagonists and primary tumor gene expression data, we sought to identify a tumor-relevant gene signature based on GR activity that would be more predictive than GR expression alone.Experimental Design: Global gene expression and GR ChIP-sequencing were performed to identify GR-regulated genes inhibited by two chemically distinct GR antagonists, mifepristone and CORT108297. Differentially expressed genes from MDA-MB-231 cells were cross-evaluated with significantly expressed genes in GR-high versus GR-low ER-negative primary breast cancers. The resulting subset of GR-targeted genes was analyzed in two independent ER-negative breast cancer cohorts to derive and then validate the GR activity signature (GRsig).Results: Gene expression pathway analysis of glucocorticoid-regulated genes (inhibited by GR antagonism) revealed cell survival and invasion functions. GR ChIP-seq analysis demonstrated that GR antagonists decreased GR chromatin association for a subset of genes. A GRsig that comprised n = 74 GR activation-associated genes (also reversed by GR antagonists) was derived from an adjuvant chemotherapy-treated Discovery cohort and found to predict probability of relapse in a separate Validation cohort (HR = 1.9; P = 0.012).Conclusions: The GRsig discovered herein identifies high-risk ER-negative/GR-positive breast cancers most likely to relapse despite administration of adjuvant chemotherapy. Because GR antagonism can reverse expression of these genes, we propose that addition of a GR antagonist to chemotherapy may improve outcome for these high-risk patients. Clin Cancer Res; 24(14); 3433-46. ©2018 AACR.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/metabolismo , Transcriptoma , Animales , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Pronóstico , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Receptores de Estrógenos/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
8.
ACS Med Chem Lett ; 8(8): 869-874, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28835804

RESUMEN

Histone lysine demethylases (KDMs) play a vital role in the regulation of chromatin-related processes. Herein, we describe our discovery of a series of potent KDM4 inhibitors that are both cell permeable and antiproliferative in cancer models. The modulation of histone H3K9me3 and H3K36me3 upon compound treatment was verified by homogeneous time-resolved fluorescence assay and by mass spectroscopy detection. Optimization of the series using structure-based drug design led to compound 6 (QC6352), a potent KDM4 family inhibitor that is efficacious in breast and colon cancer PDX models.

9.
Curr Opin Drug Discov Devel ; 7(5): 639-48, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15503866

RESUMEN

STI-571 (Gleevec) is a highly successful cancer drug due to its activity as an inhibitor of the Abelson cytoplasmic tyrosine kinase (Abl), which is constitutively active in a majority of patients with chronic myelogenous leukemia. STI-571 also inhibits two type III receptor tyrosine kinases, c-Kit and platelet-derived growth factor receptor, and functions by targeting inactive conformations of these kinases. This review focuses on recent developments in X-ray co-crystal structure analyses of STI-571 bound to Abl and the c-Kit receptor tyrosine kinase domain, and also three other relevant kinase inhibitor co-crystal structures. The similar structural features of these inactive kinases suggest they will be useful for the successful drug discovery and development of specific and targeted gene-based cancer drugs.


Asunto(s)
Conformación Molecular , Piperazinas/química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Benzamidas , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Mesilato de Imatinib , Estructura Molecular , Piperazinas/farmacología , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico
10.
Eur J Med Chem ; 61: 49-60, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22819505

RESUMEN

Focal adhesion kinase (FAK) regulates cell survival and proliferation pathways. Here we report the discovery of a highly selective series of 1,5-dihydropyrazolo[4,3-c][2,1]benzothiazines that demonstrate a novel mode of allosteric inhibition of FAK. These compounds showed slow dissociation from unphosphorylated FAK and were noncompetitive with ATP after long preincubation. Co-crystal structural analysis revealed that the compounds target a novel allosteric site within the C-lobe of the kinase domain, which induces disruption of ATP pocket formation leading to the inhibition of kinase activity. The potency of allosteric inhibition was reduced by phosphorylation of FAK. Coupled SAR analysis revealed that N-substitution of the fused pyrazole is critical to achieve allosteric binding and high selectivity among kinases.


Asunto(s)
Descubrimiento de Drogas , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Tiazinas/farmacología , Regulación Alostérica/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirazoles/síntesis química , Pirazoles/química , Relación Estructura-Actividad , Tiazinas/síntesis química , Tiazinas/química
11.
Elife ; 2: e00499, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-23580326

RESUMEN

The second-generation antiandrogen enzalutamide was recently approved for patients with castration-resistant prostate cancer. Despite its success, the duration of response is often limited. For previous antiandrogens, one mechanism of resistance is mutation of the androgen receptor (AR). To prospectively identify AR mutations that might confer resistance to enzalutamide, we performed a reporter-based mutagenesis screen and identified a novel mutation, F876L, which converted enzalutamide into an AR agonist. Ectopic expression of AR F876L rescued the growth inhibition of enzalutamide treatment. Molecular dynamics simulations performed on antiandrogen-AR complexes suggested a mechanism by which the F876L substitution alleviates antagonism through repositioning of the coactivator recruiting helix 12. This model then provided the rationale for a focused chemical screen which, based on existing antiandrogen scaffolds, identified three novel compounds that effectively antagonized AR F876L (and AR WT) to suppress the growth of prostate cancer cells resistant to enzalutamide. DOI:http://dx.doi.org/10.7554/eLife.00499.001.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Diseño de Fármacos , Mutación , Antagonistas de Andrógenos/química , Línea Celular , Resistencia a Antineoplásicos/genética , Humanos , Masculino , Simulación de Dinámica Molecular , Neoplasias de la Próstata/tratamiento farmacológico
12.
Nat Struct Mol Biol ; 15(5): 515-22, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18408731

RESUMEN

Escherichia coli endonuclease IV is an archetype for an abasic or apurinic-apyrimidinic endonuclease superfamily crucial for DNA base excision repair. Here biochemical, mutational and crystallographic characterizations reveal a three-metal ion mechanism for damage binding and incision. The 1.10-A resolution DNA-free and the 2.45-A resolution DNA-substrate complex structures capture substrate stabilization by Arg37 and reveal a distorted Zn3-ligand arrangement that reverts, after catalysis, to an ideal geometry suitable to hold rather than release cleaved DNA product. The 1.45-A resolution DNA-product complex structure shows how Tyr72 caps the active site, tunes its dielectric environment and promotes catalysis by Glu261-activated hydroxide, bound to two Zn2+ ions throughout catalysis. These structural, mutagenesis and biochemical results suggest general requirements for abasic site removal in contrast to features specific to the distinct endonuclease IV alpha-beta triose phosphate isomerase (TIM) barrel and APE1 four-layer alpha-beta folds of the apurinic-apyrimidinic endonuclease families.


Asunto(s)
Desoxirribonucleasa IV (Fago T4-Inducido)/metabolismo , Escherichia coli/enzimología , Sitios de Unión , Cristalografía por Rayos X , ADN/metabolismo , Desoxirribonucleasa IV (Fago T4-Inducido)/química , Desoxirribonucleasa IV (Fago T4-Inducido)/genética , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Estructura Secundaria de Proteína
13.
J Biol Chem ; 280(6): 4639-48, 2005 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-15513927

RESUMEN

Human 11beta-hydroxysteroid dehydrogenase type I (11beta-HSD1) is an ER-localized membrane protein that catalyzes the interconversion of cortisone and cortisol. In adipose tissue, excessive cortisol production through 11beta-HSD1 activity has been implicated in the pathogenesis of type II diabetes and obesity. We report here biophysical, kinetic, mutagenesis, and structural data on two ternary complexes of 11beta-HSD1. The combined results reveal flexible active site interactions relevant to glucocorticoid recognition and demonstrate how four 11beta-HSD1 C termini converge to form an as yet uncharacterized tetramerization motif. A C-terminal Pro-Cys motif is localized at the center of the tetramer and forms reversible enzyme disulfides that alter enzyme activity. Conformational flexibility at the tetramerization interface is coupled to structural changes at the enzyme active site suggesting how the central Pro-Cys motif may regulate enzyme activity. Together, the crystallographic and biophysical data provide a structural framework for understanding 11beta-HSD1 activities and will ultimately facilitate the development of specific inhibitors.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasas/química , Glucocorticoides/química , Secuencias de Aminoácidos , Sitios de Unión , Clonación Molecular , Cortisona/química , Cristalografía por Rayos X , Cisteína/química , Diabetes Mellitus Tipo 2/metabolismo , Dimerización , Disulfuros/química , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Escherichia coli/metabolismo , Humanos , Hidrocortisona/química , Cinética , Espectrometría de Masas , Modelos Moleculares , Mutagénesis , Obesidad/metabolismo , Oxígeno/metabolismo , Mutación Puntual , Prolina/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química
14.
Cell ; 116(1): 39-50, 2004 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-14718165

RESUMEN

Flap EndoNuclease-1 (FEN-1) and the processivity factor proliferating cell nuclear antigen (PCNA) are central to DNA replication and repair. To clarify the molecular basis of FEN-1 specificity and PCNA activation, we report here structures of FEN-1:DNA and PCNA:FEN-1-peptide complexes, along with fluorescence resonance energy transfer (FRET) and mutational results. FEN-1 binds the unpaired 3' DNA end (3' flap), opens and kinks the DNA, and promotes conformational closing of a flexible helical clamp to facilitate 5' cleavage specificity. Ordering of unstructured C-terminal regions in FEN-1 and PCNA creates an intermolecular beta sheet interface that directly links adjacent PCNA and DNA binding regions of FEN-1 and suggests how PCNA stimulates FEN-1 activity. The DNA and protein conformational changes, composite complex structures, FRET, and mutational results support enzyme-PCNA alignments and a kinked DNA pivot point that appear suitable to coordinate rotary handoffs of kinked DNA intermediates among enzymes localized by the three PCNA binding sites.


Asunto(s)
Reparación del ADN/genética , Replicación del ADN/genética , Endonucleasas de ADN Solapado/química , Endonucleasas de ADN Solapado/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Archaeoglobus , Sitios de Unión/fisiología , Dominio Catalítico/fisiología , ADN/genética , ADN/metabolismo , Sustancias Macromoleculares , Modelos Moleculares , Conformación Molecular , Antígeno Nuclear de Célula en Proliferación/química , Unión Proteica/fisiología , Estructura Secundaria de Proteína/fisiología , Estructura Terciaria de Proteína/fisiología , Saccharomyces cerevisiae , Especificidad por Sustrato/fisiología
15.
J Biol Chem ; 279(10): 8526-9, 2004 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-14672944

RESUMEN

Farnesyl pyrophosphate synthetase (FPPS) synthesizes farnesyl pyrophosphate through successive condensations of isopentyl pyrophosphate with dimethylallyl pyrophosphate and geranyl pyrophosphate. Nitrogen-containing bisphosphonate drugs used to treat osteoclast-mediated bone resorption and tumor-induced hypercalcemia are potent inhibitors of the enzyme. Here we present crystal structures of substrate and bisphosphonate complexes of FPPS. The structures reveal how enzyme conformational changes organize conserved active site residues to exploit metal-induced ionization and substrate positioning for catalysis. The structures further demonstrate how nitrogen-containing bisphosphonates mimic a carbocation intermediate to inhibit the enzyme. Together, these FPPS complexes provide a structural template for the design of novel inhibitors that may prove useful for the treatment of osteoporosis and other clinical indications including cancer.


Asunto(s)
Transferasas Alquil y Aril/química , Difosfonatos/química , Terpenos/química , Transferasas Alquil y Aril/metabolismo , Difosfonatos/metabolismo , Escherichia coli , Geraniltranstransferasa , Conformación Proteica , Relación Estructura-Actividad , Especificidad por Sustrato , Terpenos/metabolismo
16.
J Biol Chem ; 279(36): 37918-27, 2004 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-15208315

RESUMEN

Three nitric-oxide synthase (NOS) isozymes play crucial, but distinct, roles in neurotransmission, vascular homeostasis, and host defense, by catalyzing Ca(2+)/calmodulin-triggered NO synthesis. Here, we address current questions regarding NOS activity and regulation by combining mutagenesis and biochemistry with crystal structure determination of a fully assembled, electron-supplying, neuronal NOS reductase dimer. By integrating these results, we structurally elucidate the unique mechanisms for isozyme-specific regulation of electron transfer in NOS. Our discovery of the autoinhibitory helix, its placement between domains, and striking similarities with canonical calmodulin-binding motifs, support new mechanisms for NOS inhibition. NADPH, isozyme-specific residue Arg(1400), and the C-terminal tail synergistically repress NOS activity by locking the FMN binding domain in an electron-accepting position. Our analyses suggest that calmodulin binding or C-terminal tail phosphorylation frees a large scale swinging motion of the entire FMN domain to deliver electrons to the catalytic module in the holoenzyme.


Asunto(s)
Isoenzimas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Catálisis , Transporte de Electrón , Flavinas/metabolismo , Humanos , Isoenzimas/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Óxido Nítrico Sintasa/química , Óxido Nítrico Sintasa de Tipo I , Conformación Proteica , Ratas , Homología de Secuencia de Aminoácido , Difracción de Rayos X
17.
J Biol Chem ; 278(42): 41093-8, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-12882966

RESUMEN

Pasteurellosis caused by the Gram-negative pathogen Pasteurella haemolytica is a serious disease leading to death in cattle. To scavenge growth-limiting iron from the host, the pathogen utilizes the periplasmic ferric ion-binding protein A (PhFbpA) as a component of an ATP-binding cassette transport pathway. We report the 1.2-A structure of the iron-free (apo) form of PhFbpA, which is a member of the transferrin structural superfamily. The protein structure adopts a closed conformation, allowing us to reliably assign putative iron-coordinating residues. Based on our analysis, PhFbpA utilizes a unique constellation of binding site residues and anions to octahedrally coordinate an iron atom. A surprising finding in the structure is the presence of two formate anions on opposite sides of the iron-binding pocket. The formate ions tether the N- and C-terminal domains of the protein and stabilize the closed structure, also providing clues as to probable candidates for synergistic anions in the iron-loaded state. PhFbpA represents a new class of bacterial iron-binding proteins.


Asunto(s)
Proteínas Bacterianas/química , Hierro/metabolismo , Mannheimia haemolytica/metabolismo , Proteínas de la Membrana Bacteriana Externa , Proteínas Bacterianas/clasificación , Sitios de Unión , Cristalografía por Rayos X , Escherichia coli/metabolismo , Iones , Proteínas de Unión a Hierro , Modelos Moleculares , Proteínas de Unión Periplasmáticas , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Transferrina/química , Transferrina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA