Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Anal Chem ; 91(11): 7336-7345, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31045344

RESUMEN

Hydrogen-deuterium exchange mass spectrometry (HDX-MS) is an established, powerful tool for investigating protein-ligand interactions, protein folding, and protein dynamics. However, HDX-MS is still an emergent tool for quality control of biopharmaceuticals and for establishing dynamic similarity between a biosimilar and an innovator therapeutic. Because industry will conduct quality control and similarity measurements over a product lifetime and in multiple locations, an understanding of HDX-MS reproducibility is critical. To determine the reproducibility of continuous-labeling, bottom-up HDX-MS measurements, the present interlaboratory comparison project evaluated deuterium uptake data from the Fab fragment of NISTmAb reference material (PDB: 5K8A ) from 15 laboratories. Laboratories reported ∼89 800 centroid measurements for 430 proteolytic peptide sequences of the Fab fragment (∼78 900 centroids), giving ∼100% coverage, and ∼10 900 centroid measurements for 77 peptide sequences of the Fc fragment. Nearly half of peptide sequences are unique to the reporting laboratory, and only two sequences are reported by all laboratories. The majority of the laboratories (87%) exhibited centroid mass laboratory repeatability precisions of ⟨ sLab⟩ ≤ (0.15 ± 0.01) Da (1σx̅). All laboratories achieved ⟨sLab⟩ ≤ 0.4 Da. For immersions of protein at THDX = (3.6 to 25) °C and for D2O exchange times of tHDX = (30 s to 4 h) the reproducibility of back-exchange corrected, deuterium uptake measurements for the 15 laboratories is σreproducibility15 Laboratories( tHDX) = (9.0 ± 0.9) % (1σ). A nine laboratory cohort that immersed samples at THDX = 25 °C exhibited reproducibility of σreproducibility25C cohort( tHDX) = (6.5 ± 0.6) % for back-exchange corrected, deuterium uptake measurements.


Asunto(s)
Anticuerpos Monoclonales/química , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Fragmentos Fab de Inmunoglobulinas/análisis
2.
J Proteome Res ; 16(1): 355-365, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27700100

RESUMEN

Proteolysis of autoantigens can alter normal MHC class II antigen processing and has been implicated in the induction of autoimmune diseases. Many autoantigens are substrates for the protease granzyme B (GrB), but the mechanistic significance of this association is unknown. Peptidylarginine deiminase 4 (PAD4) is a frequent target of autoantibodies in patients with rheumatoid arthritis (RA) and a substrate for GrB. RA is strongly associated with specific MHC class II alleles, and elevated levels of GrB and PAD4 are found in the joints of RA patients, suggesting that GrB may alter the presentation of PAD4 by RA-associated class II alleles. In this study, complementary proteomic and immunologic approaches were utilized to define the effects of GrB cleavage on the structure, processing, and immunogenicity of PAD4. Hydrogen-deuterium exchange and a cell-free MHC class II antigen processing system revealed that proteolysis of PAD4 by GrB induced discrete structural changes in PAD4 that promoted enhanced presentation of several immunogenic peptides capable of stimulating PAD4-specific CD4+ T cells from patients with RA. This work demonstrates the existence of PAD4-specific T cells in patients with RA and supports a mechanistic role for GrB in enhancing the presentation of autoantigenic CD4+ T cell epitopes.


Asunto(s)
Artritis Reumatoide/inmunología , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Epítopos de Linfocito T/inmunología , Granzimas/inmunología , Hidrolasas/inmunología , Anciano , Secuencia de Aminoácidos , Presentación de Antígeno , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Autoanticuerpos/biosíntesis , Autoantígenos/química , Autoantígenos/genética , Sitios de Unión , Linfocitos T CD4-Positivos/patología , Estudios de Casos y Controles , Medición de Intercambio de Deuterio , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Expresión Génica , Granzimas/química , Granzimas/genética , Humanos , Hidrolasas/química , Hidrolasas/genética , Masculino , Persona de Mediana Edad , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Especificidad por Sustrato
3.
Nature ; 477(7365): 477-81, 2011 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-21892191

RESUMEN

PPARγ is the functioning receptor for the thiazolidinedione (TZD) class of antidiabetes drugs including rosiglitazone and pioglitazone. These drugs are full classical agonists for this nuclear receptor, but recent data have shown that many PPARγ-based drugs have a separate biochemical activity, blocking the obesity-linked phosphorylation of PPARγ by Cdk5. Here we describe novel synthetic compounds that have a unique mode of binding to PPARγ, completely lack classical transcriptional agonism and block the Cdk5-mediated phosphorylation in cultured adipocytes and in insulin-resistant mice. Moreover, one such compound, SR1664, has potent antidiabetic activity while not causing the fluid retention and weight gain that are serious side effects of many of the PPARγ drugs. Unlike TZDs, SR1664 also does not interfere with bone formation in culture. These data illustrate that new classes of antidiabetes drugs can be developed by specifically targeting the Cdk5-mediated phosphorylation of PPARγ.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Hipoglucemiantes/farmacología , PPAR gamma/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Líquidos Corporales/efectos de los fármacos , Células COS , Chlorocebus aethiops , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/química , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Modelos Moleculares , Obesidad/inducido químicamente , Obesidad/metabolismo , Osteogénesis/efectos de los fármacos , PPAR gamma/agonistas , PPAR gamma/química , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Rosiglitazona , Tiazolidinedionas/efectos adversos , Tiazolidinedionas/farmacología , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Aumento de Peso/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 111(45): 16154-9, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25352669

RESUMEN

Little is known regarding the identity of the population of proteins that are transported and localized to synapses. Here we describe a new approach that involves the isolation and systematic proteomic characterization of molecular motor kinesins to identify the populations of proteins transported to synapses. We used this approach to identify and compare proteins transported to synapses by kinesin (Kif) complexes Kif5C and Kif3A in the mouse hippocampus and prefrontal cortex. Approximately 40-50% of the protein cargos identified in our proteomics analysis of kinesin complexes are known synaptic proteins. We also found that the identity of kinesins and where they are expressed determine what proteins they transport. Our results reveal a previously unappreciated role of kinesins in regulating the composition of synaptic proteome.


Asunto(s)
Hipocampo/metabolismo , Cinesinas/metabolismo , Corteza Prefrontal/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Sinapsis/metabolismo , Animales , Ratones
5.
Anal Chem ; 88(12): 6607-14, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27224086

RESUMEN

Hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS) is an information-rich biophysical method for the characterization of protein dynamics. Successful applications of differential HDX-MS include the characterization of protein-ligand binding. A single differential HDX-MS data set (protein ± ligand) is often comprised of more than 40 individual HDX-MS experiments. To eliminate laborious manual processing of samples, and to minimize random and gross errors, automated systems for HDX-MS analysis have become routine in many laboratories. However, an automated system, while less prone to random errors introduced by human operators, may have systematic errors that go unnoticed without proper detection. Although the application of automated (and manual) HDX-MS has become common, there are only a handful of studies reporting the systematic evaluation of the performance of HDX-MS experiments, and no reports have been published describing a cross-site comparison of HDX-MS experiments. Here, we describe an automated HDX-MS platform that operates with a parallel, two-trap, two-column configuration that has been installed in two remote laboratories. To understand the performance of the system both within and between laboratories, we have designed and completed a test-retest repeatability study for differential HDX-MS experiments implemented at each of two laboratories, one in Florida and the other in Spain. This study provided sufficient data to do both within and between laboratory variability assessments. Initial results revealed a systematic run-order effect within one of the two systems. Therefore, the study was repeated, and this time the conclusion was that the experimental conditions were successfully replicated with minimal systematic error.


Asunto(s)
Medición de Intercambio de Deuterio/métodos , Espectrometría de Masas/métodos , Análisis de Varianza , Cromatografía Líquida de Alta Presión/instrumentación , Cromatografía Líquida de Alta Presión/métodos , Deuterio/análisis , Medición de Intercambio de Deuterio/instrumentación , Hidrógeno/análisis , Ligandos , Espectrometría de Masas/instrumentación , Péptidos/análisis , Proteínas/química , Receptores de Calcitriol/química , Reproducibilidad de los Resultados
6.
Bioorg Med Chem ; 24(4): 759-67, 2016 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-26795112

RESUMEN

Raloxifene, a selective estrogen receptor modulator (SERM), reduces fracture risk at least in part by improving the mechanical properties of bone in a cell- and estrogen receptor-independent manner. In this study, we determined that raloxifene directly interacts with the bone tissue. Through the use of multiple and complementary biophysical techniques including nuclear magnetic resonance (NMR) and Fourier transform infrared spectroscopy (FTIR), we show that raloxifene interacts specifically with the organic component or the organic/mineral composite, and not with hydroxyapatite. Structure-activity studies reveal that the basic side chain of raloxifene is an instrumental determinant in the interaction with bone. Thus, truncation of portions of the side chain reduces bone binding and also diminishes the increase in mechanical properties. Our results support a model wherein the piperidine interacts with bone matrix through electrostatic interactions with the piperidine nitrogen and through hydrophobic interactions (van der Waals) with the aliphatic groups in the side chain and the benzothiophene core. Furthermore, in silico prediction of the potential binding sites on the surface of collagen revealed the presence of a groove with sufficient space to accommodate raloxifene analogs. The hydroxyl groups on the benzothiophene nucleus, which are necessary for binding of SERMs to the estrogen receptor, are not required for binding to the bone surface, but mediate a more robust binding of the compound to the bone powder. In conclusion, we report herein a novel property of raloxifene analogs that allows them to interact with the bone tissue through potential contacts with the organic matrix and in particular collagen.


Asunto(s)
Matriz Ósea/efectos de los fármacos , Colágeno/metabolismo , Fémur/efectos de los fármacos , Clorhidrato de Raloxifeno/farmacología , Animales , Matriz Ósea/metabolismo , Colágeno/química , Perros , Durapatita/química , Fémur/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Piperidinas/química , Polilisina/química , Polilisina/metabolismo , Unión Proteica , Clorhidrato de Raloxifeno/metabolismo , Receptores de Estrógenos/metabolismo , Electricidad Estática , Relación Estructura-Actividad , Tiofenos/química
7.
Nature ; 466(7305): 451-6, 2010 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-20651683

RESUMEN

Obesity induced in mice by high-fat feeding activates the protein kinase Cdk5 (cyclin-dependent kinase 5) in adipose tissues. This results in phosphorylation of the nuclear receptor PPARgamma (peroxisome proliferator-activated receptor gamma), a dominant regulator of adipogenesis and fat cell gene expression, at serine 273. This modification of PPARgamma does not alter its adipogenic capacity, but leads to dysregulation of a large number of genes whose expression is altered in obesity, including a reduction in the expression of the insulin-sensitizing adipokine, adiponectin. The phosphorylation of PPARgamma by Cdk5 is blocked by anti-diabetic PPARgamma ligands, such as rosiglitazone and MRL24. This inhibition works both in vivo and in vitro, and is completely independent of classical receptor transcriptional agonism. Similarly, inhibition of PPARgamma phosphorylation in obese patients by rosiglitazone is very tightly associated with the anti-diabetic effects of this drug. All these findings strongly suggest that Cdk5-mediated phosphorylation of PPARgamma may be involved in the pathogenesis of insulin-resistance, and present an opportunity for development of an improved generation of anti-diabetic drugs through PPARgamma.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Diabetes Mellitus Experimental/tratamiento farmacológico , Obesidad/metabolismo , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Secuencia de Aminoácidos , Animales , Línea Celular , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Grasas de la Dieta/farmacología , Humanos , Insulina/metabolismo , Ligandos , Ratones , Modelos Moleculares , Obesidad/inducido químicamente , Obesidad/complicaciones , Obesidad/fisiopatología , PPAR gamma/agonistas , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Conformación Proteica , Rosiglitazona , Tiazolidinedionas/uso terapéutico
8.
J Biol Chem ; 289(2): 814-26, 2014 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-24187139

RESUMEN

Retinoid X receptors (RXRs) are obligate partners for several other nuclear receptors, and they play a key role in several signaling processes. Despite being a promiscuous heterodimer partner, this nuclear receptor is a target of therapeutic intervention through activation using selective RXR agonists (rexinoids). Agonist binding to RXR initiates a large conformational change in the receptor that allows for coactivator recruitment to its surface and enhanced transcription. Here we reveal the structural and dynamical changes produced when a coactivator peptide binds to the human RXRα ligand binding domain containing two clinically relevant rexinoids, Targretin and 9-cis-UAB30. Our results show that the structural changes are very similar for each rexinoid and similar to those for the pan-agonist 9-cis-retinoic acid. The four structural changes involve key residues on helix 3, helix 4, and helix 11 that move from a solvent-exposed environment to one that interacts extensively with helix 12. Hydrogen-deuterium exchange mass spectrometry reveals that the dynamics of helices 3, 11, and 12 are significantly decreased when the two rexinoids are bound to the receptor. When the pan-agonist 9-cis-retinoic acid is bound to the receptor, only the dynamics of helices 3 and 11 are reduced. The four structural changes are conserved in all x-ray structures of the RXR ligand-binding domain in the presence of agonist and coactivator peptide. They serve as hallmarks for how RXR changes conformation and dynamics in the presence of agonist and coactivator to initiate signaling.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Naftalenos/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , Receptor alfa X Retinoide/metabolismo , Tetrahidronaftalenos/metabolismo , Alitretinoína , Secuencia de Aminoácidos , Bexaroteno , Sitios de Unión , Cristalografía por Rayos X , Ácidos Grasos Insaturados/química , Humanos , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Naftalenos/química , Coactivador 2 del Receptor Nuclear/química , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor alfa X Retinoide/química , Tetrahidronaftalenos/química , Tretinoina/química , Tretinoina/metabolismo
9.
Bioorg Med Chem Lett ; 24(15): 3459-63, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24974344

RESUMEN

Modulation of the vitamin D receptor (VDR) with a ligand has the potential to be useful for the oral treatment of osteoporosis. One component of our lead generation strategy to identify synthetic ligands for VDR included a fragment based drug design approach. Screening of ligands in a VDR fluorescence polarization assay and a RXR/VDR conformation sensing assay resulted in the identification of multiple fragment hits (lean >0.30). These fragment scaffolds were subsequently evaluated for interaction with the VDR ligand binding domain using hydrogen-deuterium exchange (HDX) mass spectrometry. Significant protection of H/D exchange was observed for some fragments in helixes 3, 7, and 8 of the ligand binding domain, regions which are similar to those seen for the natural hormone VD3. The fragments appear to mimic the A-ring of VD3 thereby providing viable starting points for synthetic expansion.


Asunto(s)
Medición de Intercambio de Deuterio , Compuestos Orgánicos/farmacología , Receptores de Calcitriol/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Ligandos , Espectrometría de Masas , Modelos Moleculares , Estructura Molecular , Compuestos Orgánicos/química , Relación Estructura-Actividad
10.
Proc Natl Acad Sci U S A ; 108(46): 18678-83, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22049338

RESUMEN

Peroxisome proliferator activated receptor (PPAR) γ coactivator-1α (PGC-1α) is a potent transcriptional coactivator of oxidative metabolism and is induced in response to a variety of environmental cues. It regulates a broad array of target genes by coactivating a whole host of transcription factors. The estrogen-related receptor (ERR) family of nuclear receptors are key PGC-1α partners in the regulation of mitochondrial and tissue-specific oxidative metabolic pathways; these receptors also demonstrate strong physical and functional interactions with this coactivator. Here we perform comprehensive biochemical, biophysical, and structural analyses of the complex formed between PGC-1α and ERRγ. PGC-1α activation domain (PGC-1α(2-220)) is intrinsically disordered with limited secondary and no defined tertiary structure. Complex formation with ERRγ induces significant changes in the conformational mobility of both partners, highlighted by significant stabilization of the ligand binding domain (ERRγLBD) as determined by HDX (hydrogen/deuterium exchange) and an observed disorder-to-order transition in PGC-1α(2-220). Small-angle X-ray scattering studies allow for modeling of the solution structure of the activation domain in the absence and presence of ERRγLBD, revealing a stable and compact binary complex. These data show that PGC-1α(2-220) undergoes a large-scale conformational change when binding to the ERRγLBD, leading to substantial compaction of the activation domain. This change results in stable positioning of the N-terminal part of the activation domain of PGC-1α, favorable for assembly of an active transcriptional complex. These data also provide structural insight into the versatile coactivation profile of PGC-1α and can readily be extended to understand other transcriptional coregulators.


Asunto(s)
Proteínas de Choque Térmico/química , Receptores de Estrógenos/química , Factores de Transcripción/química , Biofisica/métodos , Humanos , Ligandos , Mitocondrias/metabolismo , Modelos Moleculares , Conformación Molecular , Oxígeno/química , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Dispersión de Radiación , Transcripción Genética , Rayos X
11.
Proc Natl Acad Sci U S A ; 108(52): 21259-64, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22160701

RESUMEN

Abscisic acid (ABA) is an essential hormone that controls plant growth, development, and responses to abiotic stresses. Central for ABA signaling is the ABA-mediated autoactivation of three monomeric Snf1-related kinases (SnRK2.2, -2.3, and -2.6). In the absence of ABA, SnRK2s are kept in an inactive state by forming physical complexes with type 2C protein phosphatases (PP2Cs). Upon relief of this inhibition, SnRK2 kinases can autoactivate through unknown mechanisms. Here, we report the crystal structures of full-length Arabidopsis thaliana SnRK2.3 and SnRK2.6 at 1.9- and 2.3-Å resolution, respectively. The structures, in combination with biochemical studies, reveal a two-step mechanism of intramolecular kinase activation that resembles the intermolecular activation of cyclin-dependent kinases. First, release of inhibition by PP2C allows the SnRK2s to become partially active because of an intramolecular stabilization of the catalytic domain by a conserved helix in the kinase regulatory domain. This stabilization enables SnRK2s to gain full activity by activation loop autophosphorylation. Autophosphorylation is more efficient in SnRK2.6, which has higher stability than SnRK2.3 and has well-structured activation loop phosphate acceptor sites that are positioned next to the catalytic site. Together, these data provide a structural framework that links ABA-mediated release of PP2C inhibition to activation of SnRK2 kinases.


Asunto(s)
Ácido Abscísico/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Modelos Moleculares , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Cristalización , Activación Enzimática , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Proteína Fosfatasa 2C , Difracción de Rayos X
13.
J Proteomics ; 302: 105198, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38777089

RESUMEN

Understanding microglial states in the aging brain has become crucial, especially with the discovery of numerous Alzheimer's disease (AD) risk and protective variants in genes such as INPP5D and TREM2, which are essential to microglia function in AD. Here we present a thorough examination of microglia-like cells and primary mouse microglia at the proteome and transcriptome levels to illuminate the roles these genes and the proteins they encode play in various cell states. First, we compared the proteome profiles of wildtype and INPP5D (SHIP1) knockout primary microglia. Our findings revealed significant proteome alterations only in the homozygous SHIP1 knockout, revealing its impact on the microglial proteome. Additionally, we compared the proteome and transcriptome profiles of commonly used in vitro microglia BV2 and HMC3 cells with primary mouse microglia. Our results demonstrated a substantial similarity between the proteome of BV2 and mouse primary cells, while notable differences were observed between BV2 and human HMC3. Lastly, we conducted targeted lipidomic analysis to quantify different phosphatidylinositols (PIs) species, which are direct SHIP1 targets, in the HMC3 and BV2 cells. This in-depth omics analysis of both mouse and human microglia enhances our systematic understanding of these microglia models. SIGNIFICANCE: Given the growing urgency of comprehending microglial function in the context of neurodegenerative diseases and the substantial therapeutic implications associated with SHIP1 modulation, we firmly believe that our study, through a rigorous and comprehensive proteomics, transcriptomics and targeted lipidomic analysis of microglia, contributes to the systematic understanding of microglial function in the context of neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Proteoma , Microglía/metabolismo , Animales , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Ratones , Proteoma/metabolismo , Proteoma/análisis , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Ratones Noqueados , Transcriptoma , Fosfatidilinositoles/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Proteómica/métodos
14.
J Biol Chem ; 287(1): 183-195, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22039047

RESUMEN

Peroxisome proliferator-activated receptors (PPARα, -ß/δ, and -γ) are a subfamily of nuclear receptors that plays key roles in glucose and lipid metabolism. PPARγ is the molecular target of the thiazolidinedione class of antidiabetic drugs that has many side effects. PPARγ is also activated by long chain unsaturated or oxidized/nitrated fatty acids, but its relationship with the medium chain fatty acids remains unclear even though the medium chain triglyceride oils have been used to control weight gain and glycemic index. Here, we show that decanoic acid (DA), a 10-carbon fatty acid and a major component of medium chain triglyceride oils, is a direct ligand of PPARγ. DA binds and partially activates PPARγ without leading to adipogenesis. Crystal structure reveals that DA occupies a novel binding site and only partially stabilizes the AF-2 helix. DA also binds weakly to PPARα and PPARß/δ. Treatments with DA and its triglyceride form improve glucose sensitivity and lipid profiles without weight gain in diabetic mice. Together, these results suggest that DA is a modulating ligand for PPARs, and the structure can aid in designing better and safer PPARγ-based drugs.


Asunto(s)
Ácidos Decanoicos/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Secuencia de Aminoácidos , Animales , Glucemia/metabolismo , Células COS , Chlorocebus aethiops , Ácidos Decanoicos/farmacología , Ácidos Decanoicos/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Diseño de Fármacos , Ligandos , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Receptores Activados del Proliferador del Peroxisoma/química , Estructura Terciaria de Proteína , Especificidad por Sustrato
15.
Nucleic Acids Res ; 39(13): 5715-28, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21415010

RESUMEN

The J-binding protein 1 (JBP1) is essential for biosynthesis and maintenance of DNA base-J (ß-d-glucosyl-hydroxymethyluracil). Base-J and JBP1 are confined to some pathogenic protozoa and are absent from higher eukaryotes, prokaryotes and viruses. We show that JBP1 recognizes J-containing DNA (J-DNA) through a 160-residue domain, DB-JBP1, with 10 000-fold preference over normal DNA. The crystal structure of DB-JBP1 revealed a helix-turn-helix variant fold, a 'helical bouquet' with a 'ribbon' helix encompassing the amino acids responsible for DNA binding. Mutation of a single residue (Asp525) in the ribbon helix abrogates specificity toward J-DNA. The same mutation renders JBP1 unable to rescue the targeted deletion of endogenous JBP1 genes in Leishmania and changes its distribution in the nucleus. Based on mutational analysis and hydrogen/deuterium-exchange mass-spectrometry data, a model of JBP1 bound to J-DNA was constructed and validated by small-angle X-ray scattering data. Our results open new possibilities for targeted prevention of J-DNA recognition as a therapeutic intervention for parasitic diseases.


Asunto(s)
Proteínas de Unión al ADN/química , ADN/química , Glucósidos/química , Proteínas Protozoarias/química , Uracilo/análogos & derivados , Secuencia de Aminoácidos , Arginina/química , Ácido Aspártico/química , Cristalografía por Rayos X , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Glucósidos/metabolismo , Lisina/química , Espectrometría de Masas , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Protozoarias/metabolismo , Dispersión del Ángulo Pequeño , Alineación de Secuencia , Uracilo/química , Uracilo/metabolismo , Difracción de Rayos X
16.
J Biol Chem ; 285(22): 16931-41, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20335173

RESUMEN

Although the family of chimaerin Rac-GAPs has recently gained significant attention for their involvement in development, cancer, and neuritogenesis, little is known about their molecular regulation. Chimaerins are activated by the lipid second messenger diacylglycerol via their C1 domain upon activation of tyrosine kinase receptors, thereby restricting the magnitude of Rac signaling in a receptor-regulated manner. Here we identified a novel regulatory mechanism for beta2-chimaerin via phosphorylation. Epidermal growth factor or the phorbol ester phorbol 12-myristate 13-acetate caused rapid phosphorylation of beta2-chimaerin on Ser(169) located in the SH2-C1 domain linker region via protein kinase Cdelta, which retained beta2-chimaerin in the cytosol and prevented its C1 domain-mediated translocation to membranes. Furthermore, despite the fact that Ser(169) phosphorylation did not alter intrinsic Rac-GAP activity in vitro, a non-phosphorylatable beta2-chimaerin mutant was highly sensitive to translocation, and displayed enhanced association with activated Rac, enhanced Rac-GAP activity, and anti-migratory properties when expressed in cells. Our results not only revealed a novel regulatory mechanism that facilitates Rac activation, but also identified a novel mechanism of cross-talk between diacylglycerol receptors that restricts beta2-chimaerin relocalization and activation.


Asunto(s)
Diglicéridos/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de Neoplasias/química , Proteína Quinasa C-delta/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Células COS , Chlorocebus aethiops , Citosol/metabolismo , Diglicéridos/química , Células HeLa , Humanos , Ratones , Mutación , Neuronas/metabolismo , Ésteres del Forbol/química , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/química , Transducción de Señal
17.
J Biol Chem ; 285(7): 5013-25, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19965867

RESUMEN

The retinoic acid receptor-related orphan receptors alpha and gamma (RORalpha (NR1F1) and RORgamma (NR1F3)) are orphan nuclear receptors and perform critical roles in regulation of development, metabolism, and immune function. Cholesterol and cholesterol sulfate have been suggested to be RORalpha ligands, but the physiological significance is unclear. To date, no endogenous RORgamma ligands have been described. Here, we demonstrate that 7-oxygenated sterols function as high affinity ligands for both RORalpha and RORgamma by directly binding to their ligand-binding domains (K(i) approximately 20 nM), modulating coactivator binding, and suppressing the transcriptional activity of the receptors. One of the 7-oxygenated sterols, 7alpha-hydroxycholesterol (7alpha-OHC), serves as a key intermediate in bile acid metabolism, and we show that 7alpha-OHC modulates the expression of ROR target genes, including Glc-6-Pase and phosphoenolpyruvate carboxykinase, in an ROR-dependent manner. Furthermore, glucose output from hepatocytes is suppressed by 7alpha-OHC functioning as an RORalpha/gamma ligand. Thus, RORalpha and RORgamma are ligand-regulated members of the NR superfamily and may serve as sensors for 7-oxygenated sterols.


Asunto(s)
Hidroxicolesteroles/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Línea Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Células Hep G2 , Humanos , Espectrometría de Masas , Ratones , Modelos Biológicos , Reacción en Cadena de la Polimerasa , Unión Proteica/fisiología
18.
Expert Rev Proteomics ; 8(1): 43-59, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21329427

RESUMEN

Functional regulation of ligand-activated receptors is driven by alterations in the conformational dynamics of the protein upon ligand binding. Differential hydrogen/deuterium exchange (HDX) coupled with mass spectrometry has emerged as a rapid and sensitive approach for characterization of perturbations in conformational dynamics of proteins following ligand binding. While this technique is sensitive to detecting ligand interactions and alterations in receptor dynamics, it also can provide important mechanistic insights into ligand regulation. For example, HDX has been used to determine a novel mechanism of ligand activation of the nuclear receptor peroxisome proliferator activated receptor-γ, perform detailed analyses of binding modes of ligands within the ligand-binding pocket of two estrogen receptor isoforms, providing insight into selectivity, and helped classify different types of estrogen receptor-α ligands by correlating their pharmacology with the way they interact with the receptor based solely on hierarchical clustering of receptor HDX signatures. Beyond small-molecule-receptor interactions, this technique has also been applied to study protein-protein complexes, such as mapping antibody-antigen interactions. In this article, we summarize the current state of the differential HDX approaches and the future outlook. We summarize how HDX analysis of protein-ligand interactions has had an impact on biology and drug discovery.


Asunto(s)
Medición de Intercambio de Deuterio/métodos , Espectrometría de Masas/métodos , Proteínas/química , Animales , Hormonas/química , Humanos , Hidrógeno/química , Ligandos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteínas Quinasas/química , Estructura Terciaria de Proteína , Receptores de Superficie Celular/química , Receptores Citoplasmáticos y Nucleares/química
20.
Int J Mass Spectrom ; 302(1-3): 59-68, 2011 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-21528013

RESUMEN

Hydrogen/deuterium exchange (HDX) mass spectrometry has been widely applied to the characterization of protein dynamics. More recently, differential HDX has been shown to be effective for the characterization of ligand binding. Previously we have described a fully automated HDX system for use as a ligand screening platform. Here we describe and validate the required data analysis workflow to facilitate the use of HDX as a robust approach for ligand screening. Following acquisition of HDX data at a single on-exchange time point (n ≥ 3), one way analysis of variance in conjunction with the Tukey multiple comparison procedure is used to establish the significance of any measured difference. Analysis results are graphed with respect to a single peptide, ligand or group of ligands, or displayed as an overview within a heat map. For the heat map display, only Δ%D values with a Tukey-adjusted P value less than 0.05 are colored. Hierarchical clustering is used to bin compounds with highly similar HDX signatures. The workflow is evaluated with a small data set showing the ligand binding domain (LDB) of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) screened against 10 functionally selective ligands. More significantly, data for the vitamin D receptor (VDR) in complex with 87 ligands are presented. To highlight the robustness and precision of our automated HDX platform we analyzed the data from 4191 replicate HDX measurements acquired over an eight month timeframe. Ninety six percent of these measurements were within 10 percent of the mean value. Work has begun to integrate these analysis and graphing components within our HDX software suite.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA