Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Proteomics ; 19(1): 16, 2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-35590248

RESUMEN

BACKGROUND: Glucagon serves as an important regulatory hormone for regulating blood glucose concentration with tight feedback control exerted by insulin and glucose. There are critical gaps in our understanding of glucagon kinetics, pancreatic α cell function and intra-islet feedback network that are disrupted in type 1 diabetes. This is important for translational research applications of evolving dual-hormone (insulin + glucagon) closed-loop artificial pancreas algorithms and their usage in type 1 diabetes. Thus, it is important to accurately measure glucagon kinetics in vivo and to develop robust models of glucose-insulin-glucagon interplay that could inform next generation of artificial pancreas algorithms. METHODS: Here, we describe the administration of novel 13C15N heavy isotope-containing glucagon tracers-FF glucagon [(Phe 6 13C9,15N; Phe 22 13C9,15N)] and FFLA glucagon [(Phe 6 13C9,15N; Phe 22 13C9,15N; Leu 14 13C6,15N; Ala 19 13C3)] followed by anti-glucagon antibody-based enrichment and LC-MS/MS based-targeted assays using high-resolution mass spectrometry to determine levels of infused glucagon in plasma samples. The optimized assay results were applied for measurement of glucagon turnover in subjects with and without type 1 diabetes infused with isotopically labeled glucagon tracers. RESULTS: The limit of quantitation was found to be 1.56 pg/ml using stable isotope-labeled glucagon as an internal standard. Intra and inter-assay variability was < 6% and < 16%, respectively, for FF glucagon while it was < 5% and < 23%, respectively, for FFLA glucagon. Further, we carried out a novel isotope dilution technique using glucagon tracers for studying glucagon kinetics in type 1 diabetes. CONCLUSIONS: The methods described in this study for simultaneous detection and quantitation of glucagon tracers have clinical utility for investigating glucagon kinetics in vivo in humans.

2.
Kidney Int ; 95(5): 1269-1272, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31010480

RESUMEN

Fibrillary glomerulonephritis (FGN) is a rare glomerular disease. Kidney biopsy is required to establish the diagnosis. Recent studies have identified abundant glomerular deposition of DNAJB9 as a unique histological marker of FGN. We developed an immunoprecipitation-based multiple reaction monitoring method to measure serum levels of DNAJB9. We detected a 4-fold higher abundance of serum DNAJB9 in FGN patients when compared to controls, including patients with other glomerular diseases. Serum DNAJB9 levels were also negatively associated with estimated glomerular filtration rate in patients with FGN. Serum DNAJB9 levels accurately predicted FGN with moderate sensitivity (67%) and with high specificity (98%) and positive and negative predictive value (89% and 95%, respectively). A receiver operating curve analysis demonstrated an AUC of 0.958. These results suggest that serum levels of DNAJB9 could be a valuable marker to predict FGN, with the potential to complement kidney biopsy for the diagnosis of FGN.


Asunto(s)
Glomerulonefritis/diagnóstico , Proteínas del Choque Térmico HSP40/sangre , Proteínas de la Membrana/sangre , Chaperonas Moleculares/sangre , Adulto , Anciano , Biomarcadores/sangre , Estudios Transversales , Diagnóstico Diferencial , Estudios de Factibilidad , Femenino , Tasa de Filtración Glomerular/fisiología , Glomerulonefritis/sangre , Glomerulonefritis/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad
3.
J Biol Chem ; 292(6): 2531-2541, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28011644

RESUMEN

Genetic models for studying localized cell suicide that halt the spread of pathogen infection and immune response activation in plants include Arabidopsis accelerated-cell-death 11 mutant (acd11). In this mutant, sphingolipid homeostasis is disrupted via depletion of ACD11, a lipid transfer protein that is specific for ceramide 1-phosphate (C1P) and phyto-C1P. The C1P binding site in ACD11 and in human ceramide-1-phosphate transfer protein (CPTP) is surrounded by cationic residues. Here, we investigated the functional regulation of ACD11 and CPTP by anionic phosphoglycerides and found that 1-palmitoyl-2-oleoyl-phosphatidic acid or 1-palmitoyl-2-oleoyl-phosphatidylglycerol (≤15 mol %) in C1P source vesicles depressed C1P intermembrane transfer. By contrast, replacement with 1-palmitoyl-2-oleoyl-phosphatidylserine stimulated C1P transfer by ACD11 and CPTP. Notably, "soluble" phosphatidylserine (dihexanoyl-phosphatidylserine) failed to stimulate C1P transfer. Also, none of the anionic phosphoglycerides affected transfer action by human glycolipid lipid transfer protein (GLTP), which is glycolipid-specific and has few cationic residues near its glycolipid binding site. These findings provide the first evidence for a potential phosphoglyceride headgroup-specific regulatory interaction site(s) existing on the surface of any GLTP-fold and delineate new differences between GLTP superfamily members that are specific for C1P versus glycolipid.


Asunto(s)
Proteínas Portadoras/metabolismo , Ceramidas/metabolismo , Fosfatidilserinas/fisiología , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Transporte Biológico , Proteínas Portadoras/química , Línea Celular , Cristalografía por Rayos X , Humanos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transferencia de Fosfolípidos , Unión Proteica , Electricidad Estática
4.
J Biol Chem ; 291(8): 3848-59, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26677223

RESUMEN

Unusually large von Willebrand factor (VWF), the first responder to vascular injury in primary hemostasis, is designed to capture platelets under the high shear stress of rheological blood flow. In type 2M von Willebrand disease, two rare mutations (G1324A and G1324S) within the platelet GPIbα binding interface of the VWF A1 domain impair the hemostatic function of VWF. We investigate structural and conformational effects of these mutations on the A1 domain's efficacy to bind collagen and adhere platelets under shear flow. These mutations enhance the thermodynamic stability, reduce the rate of unfolding, and enhance the A1 domain's resistance to limited proteolysis. Collagen binding affinity is not significantly affected indicating that the primary stabilizing effect of these mutations is to diminish the platelet binding efficiency under shear flow. The enhanced stability stems from the steric consequences of adding a side chain (G1324A) and additionally a hydrogen bond (G1324S) to His(1322) across the ß2-ß3 hairpin in the GPIbα binding interface, which restrains the conformational degrees of freedom and the overall flexibility of the native state. These studies reveal a novel rheological strategy in which the incorporation of a single glycine within the GPIbα binding interface of normal VWF enhances the probability of local unfolding that enables the A1 domain to conformationally adapt to shear flow while maintaining its overall native structure.


Asunto(s)
Mutación Missense , Desplegamiento Proteico , Factor de von Willebrand/química , Humanos , Enlace de Hidrógeno , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Reología , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
5.
J Biol Chem ; 290(50): 30053-65, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26515066

RESUMEN

Intralumenal vesicle formation of the multivesicular body is a critical step in the delivery of endocytic cargoes to the lysosome for degradation. Endosomal sorting complex required for transport III (ESCRT-III) subunits polymerize on endosomal membranes to facilitate membrane budding away from the cytoplasm to generate these intralumenal vesicles. The ATPase Vps4 remodels and disassembles ESCRT-III, but the manner in which Vps4 activity is coordinated with ESCRT-III function remains unclear. Ist1 is structurally homologous to ESCRT-III subunits and has been reported to inhibit Vps4 function despite the presence of a microtubule-interacting and trafficking domain-interacting motif (MIM) capable of stimulating Vps4 in the context of other ESCRT-III subunits. Here we report that Ist1 inhibition of Vps4 ATPase activity involves two elements in Ist1: the MIM itself and a surface containing a conserved ELYC sequence. In contrast, the MIM interaction, in concert with a more open conformation of the Ist1 core, resulted in stimulation of Vps4. Addition of the ESCRT-III subunit binding partner of Ist1, Did2, also converted Ist1 from an inhibitor to a stimulator of Vps4 ATPase activity. Finally, distinct regulation of Vps4 by Ist1 corresponded with altered ESCRT-III disassembly in vitro. Together, these data support a model in which Ist1-Did2 interactions during ESCRT-III polymerization coordinate Vps4 activity with the timing of ESCRT-III disassembly.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Mutagénesis Sitio-Dirigida , Conformación Proteica , Pliegue de Proteína , Transporte de Proteínas , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Transporte Vesicular/química
6.
J Immunol ; 190(1): 479-88, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23180824

RESUMEN

Patients with HER-2/neu-expressing breast cancer remain at risk for relapse following standard therapy. Vaccines targeting HER-2/neu to prevent relapse are in various phases of clinical testing. Many vaccines incorporate the HER-2/neu HLA-A2-binding peptide p369-377 (KIFGSLAFL), because it has been shown that CTLs specific for this epitope can directly kill HER-2/neu-overexpressing breast cancer cells. Thus, understanding how tumors process this epitope may be important for identifying those patients who would benefit from immunization. Proteasome preparations were used to determine if p369-377 was processed from larger HER-2/neu-derived fragments. HPLC, mass spectrometry, cytotoxicity assays, IFN-γ ELISPOT, and human breast cancer cell lines were used to assess the proteolytic fragments. Processing of p369-377 was not detected by purified 20S proteasome and immunoproteasome, indicating that tumor cells may not be capable of processing this Ag from the HER-2/neu protein and presenting it in the context of HLA class I. Instead, we show that other extracellular domain HER-2/neu peptide sequences are consistently processed by the proteasomes. One of these sequences, p373-382 (SLAFLPESFD), bound HLA-A2 stronger than did p369-377. CTLs specific for p373-382 recognized both p373-382 and p369-377 complexed with HLA-A2. CTLs specific for p373-382 also killed human breast cancer cell lines at higher levels than did CTLs specific for p369-377. Conversely, CTLs specific for p369-377 recognized p373-382. Peptide p373-382 is a candidate epitope for breast cancer vaccines, as it is processed by proteasomes and binds HLA-A2.


Asunto(s)
Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/metabolismo , Epítopos de Linfocito T/metabolismo , Activación de Linfocitos/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptor ErbB-2/metabolismo , Linfocitos T Citotóxicos/inmunología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Células Cultivadas , Reacciones Cruzadas/inmunología , Sistemas de Liberación de Medicamentos/métodos , Femenino , Antígeno HLA-A2/metabolismo , Humanos , Recurrencia Local de Neoplasia/enzimología , Recurrencia Local de Neoplasia/prevención & control , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/metabolismo
7.
J Proteome Res ; 10(9): 4325-33, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21755926

RESUMEN

Shotgun proteomics via mass spectrometry (MS) is a powerful technology for biomarker discovery that has the potential to lead to noninvasive disease screening mechanisms. Successful application of MS-based proteomics technologies for biomarker discovery requires accurate expectations of bias, reproducibility, variance, and the true detectable differences in platforms chosen for analyses. Characterization of the variability inherent in MS assays is vital and should affect interpretation of measurements of observed differences in biological samples. Here we describe observed biases, variance structure, and the ability to detect known differences in spike-in data sets for which true relative abundance among defined samples were known and were subsequently measured with the iTRAQ technology on two MS platforms. Global biases were observed within these data sets. Measured variability was a function of mean abundance. Fold changes were biased toward the null and variance of a fold change was a function of protein mass and abundance. The information presented herein will be valuable for experimental design and analysis of the resulting data.


Asunto(s)
Espectrometría de Masas/métodos , Fragmentos de Péptidos/análisis , Proteómica/métodos , Animales , Bovinos , Pollos , Proteínas Fúngicas/análisis , Proteínas Fúngicas/química , Caballos , Humanos , Análisis de los Mínimos Cuadrados , Fragmentos de Péptidos/química , Mapeo Peptídico , Proteínas/análisis , Proteínas/química , Proteómica/normas , Conejos , Reproducibilidad de los Resultados
8.
J Biol Chem ; 285(17): 13066-78, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20164530

RESUMEN

HET-C2 is a fungal protein that transfers glycosphingolipids between membranes and has limited sequence homology with human glycolipid transfer protein (GLTP). The human GLTP fold is unique among lipid binding/transfer proteins, defining the GLTP superfamily. Herein, GLTP fold formation by HET-C2, its glycolipid transfer specificity, and the functional role(s) of its two Trp residues have been investigated. X-ray diffraction (1.9 A) revealed a GLTP fold with all key sugar headgroup recognition residues (Asp(66), Asn(70), Lys(73), Trp(109), and His(147)) conserved and properly oriented for glycolipid binding. Far-UV CD showed secondary structure dominated by alpha-helices and a cooperative thermal unfolding transition of 49 degrees C, features consistent with a GLTP fold. Environmentally induced optical activity of Trp/Tyr/Phe (2:4:12) detected by near-UV CD was unaffected by membranes containing glycolipid but was slightly altered by membranes lacking glycolipid. Trp fluorescence was maximal at approximately 355 nm and accessible to aqueous quenchers, indicating free exposure to the aqueous milieu and consistent with surface localization of the two Trps. Interaction with membranes lacking glycolipid triggered significant decreases in Trp emission intensity but lesser than decreases induced by membranes containing glycolipid. Binding of glycolipid (confirmed by electrospray injection mass spectrometry) resulted in a blue-shifted emission wavelength maximum (approximately 6 nm) permitting determination of binding affinities. The unique positioning of Trp(208) at the HET-C2 C terminus revealed membrane-induced conformational changes that precede glycolipid uptake, whereas key differences in residues of the sugar headgroup recognition center accounted for altered glycolipid specificity and suggested evolutionary adaptation for the simpler glycosphingolipid compositions of filamentous fungi.


Asunto(s)
Proteínas Portadoras/química , Membrana Celular/química , Proteínas Fúngicas/química , Glucolípidos/química , Pliegue de Proteína , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Fluorescencia , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Glucolípidos/metabolismo , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Homología Estructural de Proteína , Triptófano/química , Triptófano/genética , Triptófano/metabolismo , Difracción de Rayos X
9.
Clin Chem ; 57(8): 1161-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21636698

RESUMEN

BACKGROUND: α-1-Antitrypsin (A1AT) deficiency results from a genetic disorder at 2 common loci. Diagnosis requires quantification of A1AT and subsequent identification of the specific variant. The current algorithm of laboratory testing for the diagnosis of A1AT deficiency uses a combination of quantification (nephelometry), genotyping, and/or phenotyping. We developed a multiple reaction monitoring liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for simultaneous quantification of A1AT and identification of the 2 most common deficiency alleles present in 95% of the patients with A1AT deficiency. METHODS: Serum samples (n = 40) were digested with trypsin, and appropriate ¹³C/¹5N-labeled standard peptides were added. We performed LC-MS/MS analysis with a 0.5- by 150-mm C18 column and H2O:acetonitrile:n-propanol:formic acid (A:98:1:1:0.2 and B:10:80:10:0.2; flow 12 µL/min) mobile phase in positive ion mode on a TSQ Quantum triple quadrupole MS system. We measured the A1AT concentration by comparison to a calibration curve and determined the phenotype by the presence or absence of variant peptides. We compared the results to the current phenotyping assay by isoelectric focusing (IEF) and the immunonephelometry quantitative assay. RESULTS: For A1AT allele detection, in 39 of 40 samples the LC-MS/MS results were identical to those obtained by IEF gel electrophoresis. The single discrepant result was rerun by IEF at a lower dilution, and the results were in concordance. The A1AT quantification by LC-MS/MS also compared favorably with nephelometry. CONCLUSIONS: The LC-MS/MS method correlates well with current phenotyping and nephelometric assays and has the potential to improve the laboratory diagnosis of genetic A1AT deficiency.


Asunto(s)
alfa 1-Antitripsina/sangre , Alelos , Cromatografía Liquida , Heterocigoto , Homocigoto , Humanos , Inmunoensayo , Focalización Isoeléctrica , Nefelometría y Turbidimetría , Fenotipo , Espectrometría de Masas en Tándem , alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/genética
10.
J Immunol ; 183(10): 6708-16, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19864598

RESUMEN

Eosinophils are multifunctional leukocytes implicated in the pathogenesis of asthma and in immunity to certain organisms. Associations between exposure to an environmental fungus, such as Alternaria, and asthma have been recognized clinically. Protease-activated receptors (PARs) are G protein-coupled receptors that are cleaved and activated by serine proteases, but their roles in innate immunity remain unknown. We previously found that human eosinophils respond vigorously to Alternaria organisms and to the secretory product(s) of Alternaria with eosinophils releasing their proinflammatory mediators. In this study, we investigated the roles of protease(s) produced by Alternaria and of PARs expressed on eosinophils in their immune responses against fungal organisms. We found that Alternaria alternata produces aspartate protease(s) and that human peripheral blood eosinophils degranulate in response to the cell-free extract of A. alternata. Eosinophils showed an increased intracellular calcium concentration in response to Alternaria that was desensitized by peptide and protease ligands for PAR-2 and inhibited by a PAR-2 antagonistic peptide. Alternaria-derived aspartate protease(s) cleaved PAR-2 to expose neo-ligands; these neo-ligands activated eosinophil degranulation in the absence of proteases. Finally, treatment of Alternaria extract with aspartate protease inhibitors, which are conventionally used for HIV-1 and other microbes, attenuated the eosinophils' responses to Alternaria. Thus, fungal aspartate protease and eosinophil PAR-2 appear critical for the eosinophils' innate immune response to certain fungi, suggesting a novel mechanism for pathologic inflammation in asthma and for host-pathogen interaction.


Asunto(s)
Alternaria/inmunología , Proteasas de Ácido Aspártico/inmunología , Neurotoxina Derivada del Eosinófilo/inmunología , Eosinófilos/inmunología , Proteínas Fúngicas/inmunología , Receptor PAR-2/inmunología , Serina Proteasas/inmunología , Alternaria/enzimología , Alternaria/metabolismo , Proteasas de Ácido Aspártico/metabolismo , Asma/inmunología , Calcio/análisis , Calcio/metabolismo , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Neurotoxina Derivada del Eosinófilo/metabolismo , Eosinófilos/efectos de los fármacos , Eosinófilos/enzimología , Eosinófilos/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Inmunidad Innata , Péptidos/farmacología , Receptor PAR-2/metabolismo , Serina Proteasas/metabolismo
11.
J Thromb Haemost ; 18(1): 79-90, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31479573

RESUMEN

BACKGROUND: A molecular basis for von Willebrand factor (VWF) self-inhibition has been proposed by which the N-terminal and C-terminal flanking sequences of the globular A1 domain disulfide loop bind to and suppress the conformational dynamics of A1. These flanking sequences are rich in O-linked glycosylation (OLG), which is known to suppress platelet adhesion to VWF, presumably by steric hindrance. The inhibitory mechanism remains unresolved as to whether inhibition is due to steric exclusion by OLGs or a direct self-association interaction that stabilizes the domain. OBJECTIVES: The platelet adhesive function, thermodynamic stability, and conformational dynamics of the wild-type and type 2M G1324S A1 domain lacking glycosylation (Escherichia coli) are compared with the wild-type glycosylated A1 domain (HEK293 cell culture) to decipher the self-inhibitory mechanism. METHODS: Surface plasmon resonance and analytical rheology are utilized to assess Glycoprotein Ibα (GPIbα) binding at equilibrium and platelet adhesion under shear flow. The conformational stability is assessed through a combination of protein unfolding thermodynamics and hydrogen-deuterium exchange mass spectrometry (HXMS). RESULTS: A1 glycosylation inhibits both GPIbα binding and platelet adhesion. Glycosylation increases the hydrodynamic size of A1 and stabilizes the thermal unfolding of A1 without changing its equilibrium stability. Glycosylation does not alter the intrinsic conformational dynamics of the A1 domain. CONCLUSIONS: These studies invalidate the proposed inhibition through conformational suppression since glycosylation within these flanking sequences does not alter the native state stability or the conformational dynamics of A1. Rather, they confirm a mechanism by which glycosylation sterically hinders platelet adhesion to the A1 domain at equilibrium and under rheological shear stress.


Asunto(s)
Complejo GPIb-IX de Glicoproteína Plaquetaria , Factor de von Willebrand , Plaquetas/metabolismo , Glicosilación , Células HEK293 , Humanos , Adhesividad Plaquetaria , Pruebas de Función Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Unión Proteica , Factor de von Willebrand/metabolismo
12.
J Mol Biol ; 432(2): 305-323, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31628947

RESUMEN

Von Willebrand factor (VWF), an exceptionally large multimeric plasma glycoprotein, functions to initiate coagulation by agglutinating platelets in the blood stream to sites of vascular injury. This primary hemostatic function is perturbed in type 2 dysfunctional subtypes of von Willebrand disease (VWD) by mutations that alter the structure and function of the platelet GPIbα adhesive VWF A1 domains. The resulting amino acid substitutions cause local disorder and misfold the native structure of the isolated platelet GPIbα-adhesive A1 domain of VWF in both gain-of-function (type 2B) and loss-of-function (type 2M) phenotypes. These structural effects have not been explicitly observed in A1 domains of VWF multimers native to blood plasma. New mass spectrometry strategies are applied to resolve the structural effects of 2B and 2M mutations in VWF to verify the presence of A1 domain structural disorder in multimeric VWF harboring type 2 VWD mutations. Limited trypsinolysis mass spectrometry (LTMS) and hydrogen-deuterium exchange mass spectrometry (HXMS) are applied to wild-type and VWD variants of the single A1, A2, and A3 domains, an A1A2A3 tridomain fragment of VWF, plasmin-cleaved dimers of VWF, multimeric recombinant VWF, and normal VWF plasma concentrates. Comparatively, these methods show that mutations known to misfold the isolated A1 domain increase the rate of trypsinolysis and the extent of hydrogen-deuterium exchange in local secondary structures of A1 within multimeric VWF. VWD mutation effects are localized to the A1 domain without appreciably affecting the structure and dynamics of other VWF domains. The intrinsic dynamics of A1 observed in recombinant fragments of VWF are conserved in plasma-derived VWF. These studies reveal that structural disorder does occur in VWD variants of the A1 domain within multimeric VWF and provides strong support for VWF misfolding as a result of some, but not all, type 2 VWD variants.


Asunto(s)
Estructura Secundaria de Proteína/genética , Deficiencias en la Proteostasis/genética , Enfermedad de von Willebrand Tipo 2/genética , Factor de von Willebrand/genética , Sustitución de Aminoácidos , Plaquetas/química , Plaquetas/metabolismo , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Mutación con Pérdida de Función/genética , Espectrometría de Masas , Dominios Proteicos/genética , Pliegue de Proteína , Multimerización de Proteína/genética , Deficiencias en la Proteostasis/sangre , Deficiencias en la Proteostasis/patología , Enfermedad de von Willebrand Tipo 2/sangre , Enfermedad de von Willebrand Tipo 2/patología , Factor de von Willebrand/química , Factor de von Willebrand/ultraestructura
13.
J Sep Sci ; 32(1): 44-56, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19051193

RESUMEN

Allergic rhinitis (AR), chronic rhinosinusitis (CRS), and asthma are prevalent airway diseases that can have a substantial impact on a patient's quality of life. MS analyses of biological fluids can effectively screen for proteins associated with disease processes, however, initial detection of diagnostic proteins is difficult due to protein complexity and dynamic range. To enhance the detection of lower abundance proteins, intact nasal lavage fluid (NLF) proteins from nonpolypoid AR and from asthmatic CRS patients were extensively fractionated prior to LC/MS/MS analysis. Pooled NLF samples were processed to remove low molecular weight molecules and high abundance plasma proteins. Anion exchange (AX) chromatography followed by RP-LC further separated the remaining intact NLF proteins. The resulting fractions were digested with trypsin and the peptides analyzed by LC/MS/MS. Spectra were searched with MASCOT, SEQUEST, and X!Tandem to obtain peptide identifications and subsequently analyzed by Scaffold software to identify parent proteins with at least 99% confidence. The 197 identified proteins are compared to those previously cited in the literature and the workflow evaluated to determine the usefulness for the detection of lower abundance proteins. This is the first extensive list of NLF proteins generated from CRS patients with coexisting asthma.


Asunto(s)
Asma , Líquido del Lavado Nasal/química , Proteínas/análisis , Rinitis , Sinusitis , Adulto , Asma/diagnóstico , Cromatografía Liquida , Enfermedad Crónica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Rinitis/diagnóstico , Sensibilidad y Especificidad , Sinusitis/diagnóstico , Espectrometría de Masas en Tándem , Factores de Tiempo
14.
Neurotoxicology ; 29(3): 444-52, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18439683

RESUMEN

PURPOSE: We present work that demonstrates that cisplatin reacts rapidly with dimethyl sulfoxide (DMSO) in solution and identify the structure and reactivity of the resulting compound. METHODS: Electrospray ionization-mass spectrometry (ESI-MS) and NMR were used to identify the chemical structure of compounds formed when DMSO reacts with cisplatin. We studied the reactivity of the identified compound with DNA. In vitro toxicity studies in neurons and cancer cells and in vivo toxicity studies in rats were used to determine both the cancer chemotherapeutic and toxic effects of the identified compound. RESULTS: Cisplatin binds rapidly with DMSO to form a DMSO adduct. The resulting compound has reduced ability to bind to double-stranded DNA both in vitro and in cells. This compound has reduced toxicity for cancer cells and neurons in vitro. In vivo nephrotoxicity studies show that the adducted compound has different nephrotoxicity and elimination characteristics than cisplatin. CONCLUSIONS: From this work, we conclude that dissolving cisplatin in DMSO results in formation of an adducted compound with different therapeutic and biological characteristics. Furthermore, future studies which propose using DMSO in combination with cisplatin for chemotherapeutic treatment in patients must be reconsidered. Due to the rapidity and nature of the reaction, DMSO and cisplatin should not be combined for patient treatment.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Cisplatino/química , Cisplatino/toxicidad , Aductos de ADN/química , Aductos de ADN/toxicidad , Dimetilsulfóxido/química , Dimetilsulfóxido/toxicidad , Síndromes de Neurotoxicidad/patología , Animales , Células Cultivadas , ADN/química , ADN/efectos de los fármacos , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , Femenino , Humanos , Leucemia L1210/metabolismo , Espectrometría de Masas , Neuronas/efectos de los fármacos , Células PC12 , Ratas , Ratas Sprague-Dawley , Soluciones , Espectrometría de Masa por Ionización de Electrospray
15.
J Am Soc Mass Spectrom ; 17(8): 1158-71, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16750384

RESUMEN

We analyzed the metal-binding properties of human centrin-2 (HsCen-2) and followed the changes in HsCen-2 structure upon metal-binding using micro-electrospray ionization mass spectrometry (muESI-MS). Apo-HsCen-2 is mostly monomeric. The ESI spectra of HsCen-2 show two charge-state distributions, representing two conformations of the protein. HsCen-2 binds four moles calcium/mol protein: one mol of calcium with high affinity, one additional mol of calcium with lower affinity, and two moles of calcium at low affinity sites. HsCen-2 binds four moles of magnesium/mol protein. The conformation giving the lower charge-state HsCen-2 by ESI, binds calcium and magnesium more readily than does the higher charge-state HsCen-2. Both conformations of HsCen-2 bind calcium more readily than magnesium. Calcium was more effective in displacing magnesium bound to HsCen-2 than vice versa. Binding of a peptide from a known binding partner, the xeroderma pigmentosum complementation group protein C (XPC), to apo-HsCen-2, occurs in the presence or the absence of calcium. Near and far-UV CD spectra of HsCen-2 show little difference with addition of calcium or magnesium. Minor changes in secondary structure are noted. Melting curves derived from temperature dependence of molar ellipticity at 222 nm for HsCen-2 show that calcium increases protein stability whereas magnesium does not. Delta 25 HsCen-2 behaves similarly to HsCen-2. We conclude that HsCen-2 binds calcium and magnesium and that calcium modulates HsCen-2 structure and function by increasing its stability without undergoing significant changes in secondary or tertiary structure.


Asunto(s)
Proteínas de Unión al Calcio/química , Proteínas de Ciclo Celular/química , Metales/química , Microquímica/métodos , Modelos Químicos , Modelos Moleculares , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrofotometría Ultravioleta/métodos , Sitios de Unión , Simulación por Computador , Unión Proteica , Conformación Proteica
17.
Clin Cancer Res ; 10(4): 1471-80, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14977851

RESUMEN

Pyrazoloacridine (PZA) is an experimental antitumor agent presently under investigation for treatment of solid tumors on the basis of its unique mechanism of action and selectivity for human solid tumor xenograft in mice. Using capillary electrophoresis coupled with electrospray ionization mass spectrometry, we have identified three oxidative PZA metabolites, 9-desmethyl-PZA, N-demethyl-PZA, and PZA N-oxide. The cytochrome p450 (CYP) isoforms involved in PZA metabolism were characterized by studies with CYP chemical inhibitors, correlation of marker activities for selected CYPs with formation of the metabolites using a human liver panel, and PZA metabolism by cDNA-expressed CYPs. 9-Desmethyl-PZA formation was catalyzed by CYP1A2, whereas N-demethyl-PZA formation was catalyzed by CYP3A4. PZA N-oxide formation was catalyzed by flavin monooxygenase (FMO) rather than CYP, as determined by studies with chemical inhibitors of FMO and metabolism by cDNA-expressed human flavin monooxygenase. After administration of [10b-(14)C]PZA to mice, six urinary metabolites were detected by high-performance liquid chromatography UV and radiochromatograms including 9-desmethyl-PZA, N-demethyl-PZA, and PZA N-oxide. Trace concentrations of 9-desmethyl-PZA and PZA N-oxide were detected in mouse plasma. PZA N-oxide and N-demethyl-PZA were detected in urine from patients after PZA administration. PZA, 9-desmethyl-PZA, and PZA N-oxide inhibited growth of A375 human melanoma cells. IC(50) values were 0.17, 0.11, and 7.0 micro M, respectively, for the three molecules.


Asunto(s)
Acridinas/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Microsomas Hepáticos/metabolismo , Oxigenasas/metabolismo , Pirazoles/farmacología , Animales , Antineoplásicos/farmacología , Catálisis , División Celular , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP3A , ADN Complementario/metabolismo , Electroforesis Capilar , Humanos , Concentración 50 Inhibidora , Cinética , Ratones , Modelos Químicos , Trasplante de Neoplasias , Oxígeno/metabolismo , Isoformas de Proteínas , Proteínas Recombinantes/química , Espectrometría de Masa por Ionización de Electrospray , Factores de Tiempo , Rayos Ultravioleta
18.
J Am Soc Mass Spectrom ; 14(6): 601-4, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12781461

RESUMEN

The advantages of the thermostable DNA polymerase from Thermococcus kodakaraensis (KOD) are demonstrated for PCR amplification with subsequent detection by mass spectrometry. Commonly used DNA polymerases for PCR amplification include those from Thermus aquaticus (Taq) and Pyrococcus furiosus (Pfu). A 116 base-pair PCR product derived from a vWA locus was amplified by Taq, Pfu, or KOD DNA polymerase and compared by agarose gel electrophoresis and electrospray ionization Fourier transform ion cyclotron resonance mass spectrometry (ESI-FTICR-MS). KOD DNA polymerase demonstrated a 2- to 3-fold increase in PCR product formation compared to Pfu or Taq, respectively, and generated blunt-ended PCR product which allows facile interpretation of the mass spectrum. Additionally, we demonstrate the advantage of using high magnetic fields to obtain unit resolution of the same 116 base pair (approximately 72 kDa) PCR product at high m/z.


Asunto(s)
ADN Polimerasa Dirigida por ADN/química , Reacción en Cadena de la Polimerasa/métodos , Thermococcus/enzimología , Análisis de Fourier , Humanos , Espectrometría de Masa por Ionización de Electrospray , Polimerasa Taq/química , Factor de von Willebrand/química
20.
Antioxid Redox Signal ; 11(5): 1097-106, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18999987

RESUMEN

We recently reported that the antineoplastic thiodioxopiperazine natural product chaetocin potently induces cellular oxidative stress, thus selectively killing cancer cells. In pursuit of underlying molecular mechanisms, we now report that chaetocin is a competitive and selective substrate for the oxidative stress mitigation enzyme thioredoxin reductase-1 (TrxR1) with lower K(m) than the TrxR1 native substrate thioredoxin (Trx; chaetocin K(m) = 4.6 +/- 0.6 microM, Trx K(m) = 104.7 +/- 26 microM), thereby attenuating reduction of the critical downstream ROS remediation substrate Trx at achieved intracellular concentrations. Consistent with a role for TrxR1 targeting in the anticancer effects of chaetocin, overexpression of the TrxR1 downstream effector Trx in HeLa cells conferred resistance to chaetocin-induced, but not to doxorubicin-induced, cytotoxicity. As the TrxR/Trx pathway is of central importance in limiting cellular reactive oxygen species (ROS)--and as chaetocin exerts its selective anticancer effects via ROS imposition--the inhibition of TrxR1 by chaetocin has potential to explain its selective anticancer effects. These observations have important implications not just with regard to the mechanism of action and clinical development of chaetocin and related thiodioxopiperazines, but also with regard to the utility of molecular targets within the thioredoxin reductase/thioredoxin pathway in the development of novel candidate antineoplastic agents.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Cromatografía Liquida , Células HeLa , Humanos , Espectrometría de Masas , Piperazinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA