Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Antimicrob Agents Chemother ; 59(2): 1052-60, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25451052

RESUMEN

Clostridium difficile infections (CDIs) are the leading cause of hospital-acquired infectious diarrhea and primarily involve two exotoxins, TcdA and TcdB. Actoxumab and bezlotoxumab are human monoclonal antibodies that neutralize the cytotoxic/cytopathic effects of TcdA and TcdB, respectively. In a phase II clinical study, the actoxumab-bezlotoxumab combination reduced the rate of CDI recurrence in patients who were also treated with standard-of-care antibiotics. However, it is not known whether the antibody combination will be effective against a broad range of C. difficile strains. As a first step toward addressing this, we tested the ability of actoxumab and bezlotoxumab to neutralize the activities of toxins from a number of clinically relevant and geographically diverse strains of C. difficile. Neutralization potencies, as measured in a cell growth/survival assay with purified toxins from various C. difficile strains, correlated well with antibody/toxin binding affinities. Actoxumab and bezlotoxumab neutralized toxins from culture supernatants of all clinical isolates tested, including multiple isolates of the BI/NAP1/027 and BK/NAP7/078 strains, at antibody concentrations well below plasma levels observed in humans. We compared the bezlotoxumab epitopes in the TcdB receptor binding domain across known TcdB sequences and found that key substitutions within the bezlotoxumab epitopes correlated with the relative differences in potencies of bezlotoxumab against TcdB of some strains, including ribotypes 027 and 078. Combined with in vitro neutralization data, epitope modeling will enhance our ability to predict the coverage of new and emerging strains by actoxumab-bezlotoxumab in the clinic.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Clostridioides difficile/efectos de los fármacos , Proteínas Bacterianas/genética , Línea Celular , Clostridioides difficile/inmunología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Epítopos/inmunología , Femenino , Humanos , Masculino
2.
J Cell Physiol ; 224(1): 273-81, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20333646

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) induces degradation of low-density lipoprotein receptor (LDLR) in the liver. It is being pursued as a therapeutic target for LDL-cholesterol reduction. Earlier genome-wide gene expression studies showed that PCSK9 over-expression in HepG2 cells resulted in up-regulation of genes in cholesterol biosynthesis and down-regulation of genes in stress response pathways; however, it was not known whether these changes were directly regulated by PCSK9 or were secondary to PCSK9-induced changes to the intracellular environment. In order to further understand the biological function of PCSK9 we treated HepG2 cells with purified recombinant wild type (WT) and D374Y gain-of-function PCSK9 proteins for 8, 24, and 48 h, and used microarray analysis to identify genome-wide expression changes and pathways. These results were compared to the changes induced by culturing HepG2 cells in cholesterol-free medium, mimicking the intracellular environment of cholesterol starvation. We determined that PCSK9-induced up-regulation of cholesterol biosynthesis genes resulted from intracellular cholesterol starvation. In addition, we identified novel pathways that are presumably regulated by PCSK9 and are independent of its effects on cholesterol uptake. These pathways included "protein ubiquitination," "xenobiotic metabolism," "cell cycle," and "inflammation and stress response." Our results indicate that PCSK9 affects metabolic pathways beyond cholesterol metabolism in HepG2 cells.


Asunto(s)
Colesterol/metabolismo , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Metabolismo de los Lípidos/genética , Neoplasias Hepáticas/genética , Serina Endopeptidasas/metabolismo , Colesterol/biosíntesis , Colesterol/deficiencia , Perfilación de la Expresión Génica/métodos , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Proproteína Convertasa 9 , Proproteína Convertasas , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/genética , Factores de Tiempo
4.
Biochem Biophys Res Commun ; 375(1): 69-73, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18675252

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to low density lipoprotein receptor (LDLR) and induces its internalization and degradation. PCSK9 binding to LDLR is mediated through the LDLR epidermal growth factor-like repeat A (EGF-A) domain. We show for the first time that an EGF-A peptide inhibits PCSK9-mediated degradation of LDLR in HepG2 cells. In addition to LDLR, we show that PCSK9 also binds directly to ApoER2 and mouse VLDLR. Importantly, binding of PCSK9 to either LDLR or mouse VLDLR was effectively inhibited by EGF-A while binding to ApoER2 was less affected. In contrast, LDL receptor-associated protein (RAP), which interacts with LDL receptor repeat type A (LA) domains, inhibited PCSK9 binding to ApoER2 with greater efficacy than either LDLR or mVLDLR. These data demonstrate that while PCSK9 binds several receptors via its EGF-A binding domain, additional contacts with other receptor domains are also involved.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Receptores de LDL/metabolismo , Receptores de Lipoproteína/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Línea Celular , Humanos , Proteínas Relacionadas con Receptor de LDL , Ratones , Péptidos/metabolismo , Proproteína Convertasa 9 , Proproteína Convertasas , Estructura Terciaria de Proteína , Serina Endopeptidasas/genética
5.
Expert Rev Mol Diagn ; 8(2): 179-87, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18366304

RESUMEN

KCa3.1 is a calcium-activated intermediate-conductance potassium ion channel. In humans the channel is expressed in several secretory organs and subtypes of hematopoietic cells, but not detected in excitable tissues. The mRNA level for KCa3.1 is upregulated in activated leukocytes, mitogen-induced endothelial cells and vascular smooth muscle cells, and several types of human cancers, suggesting a possible role for the channel in inflammatory and oncology diseases. Several potent and selective KCa3.1 blockers, including clotrimazole and its analogs TRAM-34 and ICA-17043, have been used to investigate the involvement of the channel in human disease. The compounds have been shown to suppress the proliferation of several cancer cells in vitro and the growth of the corresponding cancers in vivo, consistent with an oncologic indication. TRAM-34 also ameliorates symptoms in experimental autoimmune encephalomyelitis and several models of cardiovascular diseases, arguing for a role of the channel in inflammatory diseases. These results suggest several important opportunities for therapeutics based on KCa3.1. Further efforts will establish the optimal indication for these ion channel inhibitors.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Encefalomielitis Autoinmune Experimental/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neoplasias/metabolismo , Vasculitis/metabolismo , Animales , Biomarcadores de Tumor/antagonistas & inhibidores , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Endotelio Vascular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Mitógenos/farmacología , Músculo Liso Vascular/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Especificidad de Órganos/efectos de los fármacos , ARN Mensajero/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Vasculitis/tratamiento farmacológico
6.
J Med Chem ; 51(3): 581-8, 2008 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-18198821

RESUMEN

Melanin-concentrating hormone receptor 1 (MCH-R1) is a G-protein-coupled receptor (GPCR) and a target for the development of therapeutics for obesity. The structure-based development of MCH-R1 and other GPCR antagonists is hampered by the lack of an available experimentally determined atomic structure. A ligand-steered homology modeling approach has been developed (where information about existing ligands is used explicitly to shape and optimize the binding site) followed by docking-based virtual screening. Top scoring compounds identified virtually were tested experimentally in an MCH-R1 competitive binding assay, and six novel chemotypes as low micromolar affinity antagonist "hits" were identified. This success rate is more than a 10-fold improvement over random high-throughput screening, which supports our ligand-steered method. Clearly, the ligand-steered homology modeling method reduces the uncertainty of structure modeling for difficult targets like GPCRs.


Asunto(s)
Ligandos , Modelos Moleculares , Receptores de la Hormona Hipofisaria/antagonistas & inhibidores , Receptores de la Hormona Hipofisaria/química , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/química , Animales , Sitios de Unión , Unión Competitiva , Células CHO , Bovinos , Cricetinae , Cricetulus , Bases de Datos Factuales , Humanos , Receptores de la Hormona Hipofisaria/metabolismo , Receptores de Somatostatina/metabolismo , Rodopsina/química , Homología de Secuencia de Aminoácido , Procesos Estocásticos , Relación Estructura-Actividad , Termodinámica
7.
Genomics ; 90(5): 629-35, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17869477

RESUMEN

Mice lacking GPR103A expression display osteopenia. Analysis of mouse quantitative trait loci literature associated with bone mineral density suggested GPR103A ligand P518/Qrfp (chromosome 2qB) as a candidate osteoporosis gene. Promoter and coding regions of mouse P518/Qrfp were sequenced from genomic DNA obtained from the osteoporosis-prone strain SAMP6 and control strains SAMR1, A/J, AKR/J, BALB/c, C3H/HeJ, C57BL/6J, and DBA/2J. Four single-nucleotide polymorphisms (SNPs) were identified in only SAMP6 genomic DNA, g.-1773 T-->C, g.110 A-->G (N37S), g.188 G-->A (R63K), and g.135 T-->C (H45H). The promoter SNP generated a novel neuron-restrictive silencing factor binding site, a repressor that decreases gene expression in nonneuronal tissues. TaqMan analysis demonstrated fivefold lower P518/Qrfp liver expression in SAMP6 versus SAMR1 or C57BL/6J control strains. Tissue distribution of human, mouse, and rat P518/Qrfp and its receptors showed expression in bone and spinal cord. A direct role for P518/Qrfp function in maintaining bone mineral density is suggested.


Asunto(s)
Enfermedades Óseas Metabólicas/genética , Sistemas de Lectura Abierta/genética , Péptidos/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Carácter Cuantitativo Heredable , Receptores Acoplados a Proteínas G/genética , Secuencia de Aminoácidos , Animales , Densidad Ósea , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ligandos , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Ratas , Homología de Secuencia de Aminoácido , Distribución Tisular
8.
Biochem J ; 398(3): 423-30, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16724960

RESUMEN

The Gpbar1 [G-protein-coupled BA (bile acid) receptor 1] is a recently identified cell-surface receptor that can bind and is activated by BAs, but its physiological role is unclear. Using targeted deletion of the Gpbar1 gene in mice, we show that the gene plays a critical role in the maintenance of bile lipid homoeostasis. Mice lacking Gpbar1 expression were viable, developed normally and did not show significant difference in the levels of cholesterol, BAs or any other bile constituents. However, they did not form cholesterol gallstones when fed a cholic acid-containing high-fat diet, and liver-specific gene expression indicated that Gpbar1-deficient mice have altered feedback regulation of BA synthesis. These results suggest that Gpbar1 plays a critical role in the formation of gallstones, possibly via a regulatory mechanism involving the cholesterol 7alpha-hydroxylase pathway.


Asunto(s)
Colesterol/análisis , Cálculos Biliares/genética , Cálculos Biliares/metabolismo , Eliminación de Gen , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Ácidos y Sales Biliares/biosíntesis , Colesterol 7-alfa-Hidroxilasa/metabolismo , Grasas de la Dieta/metabolismo , Vesícula Biliar/patología , Cálculos Biliares/química , Regulación de la Expresión Génica , Hígado/patología , Ratones , Ratones Noqueados , ARN Mensajero
9.
J Virol ; 78(8): 4134-44, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047829

RESUMEN

AD101 and SCH-C are two chemically related small molecules that inhibit the entry of human immunodeficiency virus type 1 (HIV-1) via human CCR5. AD101 also inhibits HIV-1 entry via rhesus macaque CCR5, but SCH-C does not. Among the eight residues that differ between the human and macaque versions of the coreceptor, only one, methionine-198, accounts for the insensitivity of macaque CCR5 to inhibition by SCH-C. Thus, the macaque coreceptor engineered to contain the natural human CCR5 residue (isoleucine) at position 198 is sensitive to HIV-1 entry inhibition by SCH-C, whereas a human CCR5 mutant containing the corresponding macaque residue (methionine) is resistant. Position 198 is in CCR5 transmembrane (TM) helix 5 and is not located within the previously defined binding site for AD101 and SCH-C, which involves residues in TM helices 1, 2, 3, and 7. SCH-C binds to human CCR5 whether residue 198 is isoleucine or methionine, and it also binds to macaque CCR5. However, the binding of a conformation-dependent monoclonal antibody to human CCR5 is inhibited by SCH-C only when residue 198 is isoleucine. These observations, taken together, suggest that the antiviral effects of SCH-C and AD101 involve stabilization, or induction, of a CCR5 conformation that is not compatible with HIV-1 infection. However, SCH-C is unable to exert this effect on CCR5 conformation when residue 198 is methionine. The region of CCR5 near residue 198 has, therefore, an important influence on the conformational state of this receptor.


Asunto(s)
Antagonistas de los Receptores CCR5 , VIH-1/efectos de los fármacos , Piperidinas , Receptores CCR5/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Línea Celular , Quimiocina CCL5/antagonistas & inhibidores , Óxidos N-Cíclicos/farmacología , VIH-1/patogenicidad , Humanos , Macaca mulatta , Modelos Biológicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Oximas , Conformación Proteica , Estructura Terciaria de Proteína , Piridinas/farmacología , Receptores CCR5/química , Transducción de Señal/efectos de los fármacos , Especificidad de la Especie
10.
Mamm Genome ; 14(5): 341-9, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12856286

RESUMEN

UNLABELLED: PepPat, a hybrid method that combines pattern matching with similarity scoring, is described. We also report PepPat's application in the identification of a novel tachykinin-like peptide. PepPat takes as input a query peptide and a user-specified regular expression pattern within the peptide. It first performs a database pattern match and then ranks candidates on the basis of their similarity to the query peptide. PepPat calculates similarity over the pattern spanning region, enhancing PepPat's sensitivity for short query peptides. PepPat can also search for a user-specified number of occurrences of a repeated pattern within the target sequence. We illustrate PepPat's application in short peptide ligand mining. As a validation example, we report the identification of a novel tachykinin-like peptide, C14TKL-1, and show it is an NK1 (neuokinin receptor 1) agonist whose message is widely expressed in human periphery. AVAILABILITY: PepPat is offered online at: http://peppat.cbi.pku.edu.cn.


Asunto(s)
Técnicas Genéticas , Oligopéptidos/genética , Receptores de Neuroquinina-1/genética , Taquicininas/genética , Algoritmos , Cartilla de ADN/química , Bases de Datos Factuales , Humanos , Oligopéptidos/metabolismo , Reacción en Cadena de la Polimerasa , Receptores de Neuroquinina-1/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Taquicininas/metabolismo
11.
Artículo en Chino | MEDLINE | ID: mdl-12417917

RESUMEN

The iron response element (IRE) is a highly conserved RNA stem loop structure. It is the binding site of iron regulatory protein (IRP). IRP binding to IRE is regulated by cellular iron. When cells are derived of iron, IRP binds IRE. If IRE is located at 5'UTR, IRP binding will inhibit translation initiation, else if IRE is at 3'UTR, IRP binding will stabilize mRNA and prevent it from degradation. So far all known IREs have C at the 1 position and G at the 5 position of the loop (C1G5 type). In vitro studies suggest that the U1A5 type IRE, which has U and A at the 1 and 5 loop position respectively, binds well to IRP. However, U1A5 type's in vivo existence is still elusive. IRE-IRP binding is involved in the regulation of iron metabolism, oxidative stress and possibly aging. Here we use an improved computation method performing a comprehensive search of IRE in human and mouse genes. We try to catalog potential human and mouse IRE containing genes, at the same time identify potential U1A5 IREs.


Asunto(s)
Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/genética , Hierro/farmacología , Elementos de Respuesta/genética , Programas Informáticos , Regiones no Traducidas 3'/química , Regiones no Traducidas 5'/química , Animales , Secuencia de Bases , Sitios de Unión/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hierro/metabolismo , Proteínas de Unión a Hierro/metabolismo , Ratones , Conformación de Ácido Nucleico , Elementos de Respuesta/efectos de los fármacos
12.
Mol Pharmacol ; 62(1): 38-47, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12065753

RESUMEN

The histamine H4 receptor is a novel G-protein-coupled receptor with a unique pharmacological profile. The distribution of H4 mRNA suggests that it may play a role in the regulation of immune function, particularly with respect to allergy and asthma. To define the histamine-binding site of this receptor, molecular modeling and site-directed mutagenesis were used to predict and alter amino acids residing in the histamine-binding pocket. The effects of these alterations on histamine binding and receptor activation were then assessed. Our results indicate that Asp94 (3.32) in transmembrane region (TM) 3 and Glu182 (5.46) in TM5 are critically involved in histamine binding. Asp94 probably serves as a counter-anion to the cationic amino group of histamine, whereas Glu182 (5.46) interacts with the N(tau) nitrogen atom of the histamine imidazole ring via an ion pair. In contrast, Thr178 (5.42) and Ser179 (5.43) in TM5 are not significantly involved in either histamine binding or receptor activation. These results resemble those for the analogous residues in the H1 histamine receptor but contrast with findings regarding the H2 histamine receptor. Our results also demonstrate that Asn147 (4.57) in TM4 and Ser320 (6.52) in TM6 play a role in receptor activation but are not involved in histamine binding. Taken together, these data indicate that although histamine seems to bind to the H4 receptor in a fashion similar to that predicted for the other histamine receptor subtypes, there are also important differences that can probably be exploited for the discovery of novel H4-selective compounds.


Asunto(s)
Histamina/metabolismo , Receptores Acoplados a Proteínas G , Receptores Histamínicos/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Asparagina/genética , Sitios de Unión , Calcio/metabolismo , Células Cultivadas , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fenilalanina/genética , Receptores Histamínicos/química , Receptores Histamínicos/genética , Receptores Histamínicos H4 , Homología de Secuencia de Aminoácido , Serina/genética , Tirosina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA