Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mo Med ; 110(4): 325-30, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24003651

RESUMEN

The Cho and Baldan labs focus their efforts on novel pathways that control atherogenesis. MIF (Macrophage migration inhibitory factor) recruits macrophages to atherosclerotic lesions and activates the production of matrix proteinases, which in turn destabilize atherosclerotic plaques. On the other hand, miR-33 coordinates the expression of several sterol transporters essential for high-density lipoprotein metabolism and bile secretion. Thus, both MIF and miR-33 are promising therapeutic targets to manage patients at risk of developing atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , MicroARNs/metabolismo , Aterosclerosis/genética , Biomarcadores/metabolismo , HDL-Colesterol/metabolismo , Humanos
2.
Chem Biol ; 18(9): 1089-101, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21944748

RESUMEN

The screening of bioactive compound libraries can be an effective approach for repositioning FDA-approved drugs or discovering new pharmacophores. Hookworms are blood-feeding, intestinal nematode parasites that infect up to 600 million people worldwide. Vaccination with recombinant Ancylostoma ceylanicum macrophage migration inhibitory factor (rAceMIF) provided partial protection from disease, thus establishing a "proof-of-concept" for targeting AceMIF to prevent or treat infection. A high-throughput screen (HTS) against rAceMIF identified six AceMIF-specific inhibitors. A nonsteroidal anti-inflammatory drug (NSAID), sodium meclofenamate, could be tested in an animal model to assess the therapeutic efficacy in treating hookworm disease. Furosemide, an FDA-approved diuretic, exhibited submicromolar inhibition of rAceMIF tautomerase activity. Structure-activity relationships of a pharmacophore based on furosemide included one analog that binds similarly to the active site, yet does not inhibit the Na-K-Cl symporter (NKCC1) responsible for diuretic activity.


Asunto(s)
Ancylostomatoidea/metabolismo , Reposicionamiento de Medicamentos , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Ancylostomatoidea/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Diuréticos/química , Diuréticos/farmacología , Diuréticos/uso terapéutico , Furosemida/química , Furosemida/farmacología , Furosemida/uso terapéutico , Ensayos Analíticos de Alto Rendimiento , Infecciones por Uncinaria/tratamiento farmacológico , Humanos , Cinética , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ácido Meclofenámico/química , Ácido Meclofenámico/farmacología , Ácido Meclofenámico/uso terapéutico , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico
3.
Science ; 331(6018): 768-72, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21311022

RESUMEN

Endocrine tumors such as aldosterone-producing adrenal adenomas (APAs), a cause of severe hypertension, feature constitutive hormone production and unrestrained cell proliferation; the mechanisms linking these events are unknown. We identify two recurrent somatic mutations in and near the selectivity filter of the potassium (K(+)) channel KCNJ5 that are present in 8 of 22 human APAs studied. Both produce increased sodium (Na(+)) conductance and cell depolarization, which in adrenal glomerulosa cells produces calcium (Ca(2+)) entry, the signal for aldosterone production and cell proliferation. Similarly, we identify an inherited KCNJ5 mutation that produces increased Na(+) conductance in a Mendelian form of severe aldosteronism and massive bilateral adrenal hyperplasia. These findings explain pathogenesis in a subset of patients with severe hypertension and implicate loss of K(+) channel selectivity in constitutive cell proliferation and hormone production.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Adenoma Corticosuprarrenal/genética , Aldosterona/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Hiperaldosteronismo/genética , Hipertensión/genética , Mutación , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Glándulas Suprarrenales/patología , Adenoma Corticosuprarrenal/metabolismo , Adenoma Corticosuprarrenal/patología , Línea Celular , Proliferación Celular , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/química , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Humanos , Hiperaldosteronismo/metabolismo , Hiperaldosteronismo/patología , Hiperplasia , Hipertensión/metabolismo , Masculino , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Potasio/metabolismo , Multimerización de Proteína , Sodio/metabolismo , Zona Glomerular/metabolismo , Zona Glomerular/patología
5.
Proc Natl Acad Sci U S A ; 107(25): 11313-8, 2010 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-20534506

RESUMEN

AV411 (ibudilast; 3-isobutyryl-2-isopropylpyrazolo-[1,5-a]pyridine) is an antiinflammatory drug that was initially developed for the treatment of bronchial asthma but which also has been used for cerebrovascular and ocular indications. It is a nonselective inhibitor of various phosphodiesterases (PDEs) and has varied antiinflammatory activity. More recently, AV411 has been studied as a possible therapeutic for the treatment of neuropathic pain and opioid withdrawal through its actions on glial cells. As described herein, the PDE inhibitor AV411 and its PDE-inhibition-compromised analog AV1013 inhibit the catalytic and chemotactic functions of the proinflammatory protein, macrophage migration inhibitory factor (MIF). Enzymatic analysis indicates that these compounds are noncompetitive inhibitors of the p-hydroxyphenylpyruvate (HPP) tautomerase activity of MIF and an allosteric binding site of AV411 and AV1013 is detected by NMR. The allosteric inhibition mechanism is further elucidated by X-ray crystallography based on the MIF/AV1013 binary and MIF/AV1013/HPP ternary complexes. In addition, our antibody experiments directed against MIF receptors indicate that CXCR2 is the major receptor for MIF-mediated chemotaxis of peripheral blood mononuclear cells.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/metabolismo , Piridinas/química , Sitio Alostérico , Sitios de Unión , Catálisis , Quimiotaxis , Cristalografía por Rayos X/métodos , Citocinas/metabolismo , Humanos , Inflamación , Oxidorreductasas Intramoleculares/química , Cinética , Ácidos Fenilpirúvicos/química , Inhibidores de Agregación Plaquetaria/farmacología , Unión Proteica , Espectrometría de Fluorescencia/métodos
6.
J Med Chem ; 51(19): 5984-92, 2008 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-18778048

RESUMEN

HisG is an ATP-phosphoribosyl transferase (ATPPRTase) that catalyzes the first step in the biosynthetic pathway for histidine. Among the enzymes in this pathway, only HisG represents a potential drug target for tuberculosis. Only a few inhibitors with limited potency for HisG are currently known. To discover more potent and diverse inhibitors, virtual screening was performed. The crystal structure of M. tuberculosis HisG has been solved and reveals a large, solvent-exposed active site with subsites for ATP and PRPP substrates. Two docking algorithms, GOLD and FLEXX, were used to screen two large libraries, Chembridge and NCI, containing over 500000 compounds combined. An initial subset of top-ranked compounds were selected and assayed, and seven were found to have enzyme inhibition activity at micromolar concentrations. Several of the hits contained a nitrobenzothiazole fragment, which was predicted to dock into the monophosphate-binding loop, and this binding mode was confirmed by crystallographic evidence. A secondary screen was performed to identify compounds with similar structures. Several of these also exhibited micromolar inhibition. Furthermore, two of the compounds showed bacteriocidal activity in a whole-cell assay against Mycobacterium smegmatis.


Asunto(s)
ATP Fosforribosil Transferasa/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Benzotiazoles/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Algoritmos , Benzotiazoles/síntesis química , Benzotiazoles/química , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Mycobacterium smegmatis/crecimiento & desarrollo , Bibliotecas de Moléculas Pequeñas , Estereoisomerismo , Relación Estructura-Actividad , Propiedades de Superficie
7.
Trends Parasitol ; 24(8): 355-63, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18603473

RESUMEN

Chronic helminth infections are associated with modulation of host cellular immune responses, presumably to prolong parasite survival within the mammalian host. This phenomenon is attributed, at least in part, to the elaboration of parasite molecules, including orthologs of host cytokines and receptors, at the host-parasite interface. This review describes recent progress in the characterization of macrophage migration inhibitory factor (MIF) orthologs from parasitic nematodes. The roles of these molecules in parasite developmental biology and pathogenesis are discussed. Further knowledge of the species-specific activities and three-dimensional structures of human and parasitic nematode MIF molecules should make them ideal targets for drug- and/or vaccine-based strategies aimed at nematode disease control.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Nematodos/metabolismo , Infecciones por Nematodos/inmunología , Animales , Regulación de la Expresión Génica/fisiología , Interacciones Huésped-Parásitos/inmunología , Humanos , Factores Inhibidores de la Migración de Macrófagos/genética , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Nematodos/genética , Nematodos/inmunología , Infecciones por Nematodos/parasitología
8.
J Immunol ; 180(12): 8250-61, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18523291

RESUMEN

Parasitic organisms have evolved specialized strategies to evade immune defense mechanisms. We describe herein an ortholog of the cytokine, macrophage migration inhibitory factor (MIF), which is produced by the obligate intracellular parasite, Leishmania major. The Leishmania MIF protein, Lm1740MIF, shows significant structural homology with human MIF as revealed by a high-resolution x-ray crystal structure (1.03 A). Differences between the two proteins in the N-terminal tautomerization site are evident, and we provide evidence for the selective, species-specific inhibition of MIF by small-molecule antagonists that target this site. Lm1740MIF shows significant binding interaction with the MIF receptor, CD74 (K(d) = 2.9 x 10(-8) M). Like its mammalian counterpart, Lm1740MIF induces ERK1/2 MAP kinase activation in a CD74-dependent manner and inhibits the activation-induced apoptosis of macrophages. The ability of Lm1740MIF to inhibit apoptosis may facilitate the persistence of Leishmania within the macrophage and contribute to its evasion from immune destruction.


Asunto(s)
Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/fisiología , Leishmania major/química , Leishmania major/inmunología , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/fisiología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/parasitología , Homología Estructural de Proteína , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/metabolismo , Antígenos de Diferenciación de Linfocitos B/fisiología , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/fisiología , Línea Celular , Células Cultivadas , Cristalografía por Rayos X , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Antígenos de Histocompatibilidad Clase II/fisiología , Humanos , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Leishmania major/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Macrófagos Peritoneales/enzimología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
9.
J Neurosci ; 28(4): 932-43, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18216201

RESUMEN

At most excitatory central synapses, glutamate is released from presynaptic terminals and binds to postsynaptic AMPA receptors, initiating a series of conformational changes that result in ion channel opening. Efficient transmission at these synapses requires that glutamate binding to AMPA receptors results in rapid and near-synchronous opening of postsynaptic receptor channels. In addition, if the information encoded in the frequency of action potential discharge is to be transmitted faithfully, glutamate must dissociate from the receptor quickly, enabling the synapse to discriminate presynaptic action potentials that are spaced closely in time. The current view is that the efficacy of agonists is directly related to the extent to which ligand binding results in closure of the binding domain. For glutamate to dissociate from the receptor, however, the binding domain must open. Previously, we showed that mutations in glutamate receptor subunit 2 that should destabilize the closed conformation not only sped deactivation but also altered the relative efficacy of glutamate and quisqualate. Here we present x-ray crystallographic and single-channel data that support the conclusions that binding domain closing necessarily precedes channel opening and that the kinetics of conformational changes at the level of the binding domain importantly influence ion channel gating. Our findings suggest that the stability of the closed-cleft conformation has been tuned during evolution so that glutamate dissociates from the receptor as rapidly as possible but remains an efficacious agonist.


Asunto(s)
Activación del Canal Iónico/fisiología , Receptores AMPA/química , Receptores AMPA/metabolismo , Línea Celular , Activación del Canal Iónico/genética , Ligandos , Mutación , Unión Proteica/genética , Estructura Secundaria de Proteína/genética , Receptores AMPA/genética , Factores de Tiempo
10.
J Biol Chem ; 282(32): 23447-56, 2007 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-17567581

RESUMEN

Hookworms, parasitic nematodes that infect nearly one billion people worldwide, are a major cause of anemia and malnutrition. We hypothesize that hookworms actively manipulate the host immune response through the production of specific molecules designed to facilitate infection by larval stages and adult worm survival within the intestine. A full-length cDNA encoding a secreted orthologue of the human cytokine, Macrophage Migration Inhibitory Factor (MIF) has been cloned from the hookworm Ancylostoma ceylanicum. Elucidation of the three-dimensional crystal structure of recombinant AceMIF (rAceMIF) revealed an overall structural homology with significant differences in the tautomerase sites of the human and hookworm proteins. The relative bioactivities of human and hookworm MIF proteins were compared using in vitro assays of tautomerase activity, macrophage migration, and binding to MIF receptor CD74. The activity of rAceMIF was not inhibited by the ligand ISO-1, which was previously determined to be an inhibitor of the catalytic site of human MIF. These data define unique immunological, structural, and functional characteristics of AceMIF, thereby establishing the potential for selectively inhibiting the hookworm cytokine as a means of reducing parasite survival and disease pathogenesis.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/química , Antígenos de Diferenciación de Linfocitos B/fisiología , Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase II/fisiología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Secuencia de Aminoácidos , Ancylostoma , Animales , Cricetinae , Citocinas/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mesocricetus , Datos de Secuencia Molecular , Unión Proteica , Conejos , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido
11.
J Biol Chem ; 282(13): 10018-10027, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17264079

RESUMEN

CXCL12 (SDF-1alpha) and CXCR4 are critical for embryonic development and cellular migration in adults. These proteins are involved in HIV-1 infection, cancer metastasis, and WHIM disease. Sequestration and presentation of CXCL12 to CXCR4 by glycosaminoglycans (GAGs) is proposed to be important for receptor activation. Mutagenesis has identified CXCL12 residues that bind to heparin. However, the molecular details of this interaction have not yet been determined. Here we demonstrate that soluble heparin and heparan sulfate negatively affect CXCL12-mediated in vitro chemotaxis. We also show that a cluster of basic residues in the dimer interface is required for chemotaxis and is a target for inhibition by heparin. We present structural evidence for binding of an unsaturated heparin disaccharide to CXCL12 attained through solution NMR spectroscopy and x-ray crystallography. Increasing concentrations of the disaccharide altered the two-dimensional (1)H-(15)N-HSQC spectra of CXCL12, which identified two clusters of residues. One cluster corresponds to beta-strands in the dimer interface. The second includes the amino-terminal loop and the alpha-helix. In the x-ray structure two unsaturated disaccharides are present. One is in the dimer interface with direct contacts between residues His(25), Lys(27), and Arg(41) of CXCL12 and the heparin disaccharide. The second disaccharide contacts Ala(20), Arg(21), Asn(30), and Lys(64). This is the first x-ray structure of a CXC class chemokine in complex with glycosaminoglycans. Based on the observation of two heparin binding sites, we propose a mechanism in which GAGs bind around CXCL12 dimers as they sequester and present CXCL12 to CXCR4.


Asunto(s)
Quimiocinas CXC/química , Quimiocinas CXC/fisiología , Heparina/química , Heparina/fisiología , Línea Celular Tumoral , Quimiocina CXCL12 , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Quimiotaxis , Cristalografía por Rayos X , Dimerización , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Heparina/metabolismo , Humanos , Unión Proteica/fisiología , Receptores CXCR4/metabolismo
13.
J Biol Chem ; 278(10): 8333-9, 2003 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-12511575

RESUMEN

The N-1-(5'-phosphoribosyl)-ATP transferase catalyzes the first step of the histidine biosynthetic pathway and is regulated by a feedback mechanism by the product histidine. The crystal structures of the N-1-(5'-phosphoribosyl)-ATP transferase from Mycobacterium tuberculosis in complex with inhibitor histidine and AMP has been determined to 1.8 A resolution and without ligands to 2.7 A resolution. The active enzyme exists primarily as a dimer, and the histidine-inhibited form is a hexamer. The structure represents a new fold for a phosphoribosyltransferase, consisting of three continuous domains. The inhibitor AMP binds in the active site cavity formed between the two catalytic domains. A model for the mechanism of allosteric inhibition has been derived from conformational differences between the AMP:His-bound and apo structures.


Asunto(s)
ATP Fosforribosil Transferasa/química , Mycobacterium tuberculosis/enzimología , ATP Fosforribosil Transferasa/genética , ATP Fosforribosil Transferasa/metabolismo , Adenosina Monofosfato/metabolismo , Dominio Catalítico , Clonación Molecular , Modelos Moleculares , Mycobacterium tuberculosis/genética , Estructura Cuaternaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA