Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nanoscale ; 16(11): 5715-5728, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38407269

RESUMEN

Inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are chronic inflammatory diseases of the gastrointestinal tract and are characterized by chronic recurrent ulceration of the bowels. Colon-targeted drug delivery systems (DDS) have received significant attention for their potential to treat IBD by improving the inflamed tissue selectivity. Herein, antiMUC5AC-decorated drug loaded nanoparticles (NP) are suggested for active epithelial targeting and selective adhesion to the inflamed tissue in experimental colitis. NPs conjugated with antiMUC5AC (anti-MUC5) were tested for their degree of bioadhesion with HT29-MTX cells by comparison with non-targeted BSA-NP conjugates. In vivo, the selectivity of bioadhesion and the influence of ligand density in bioadhesion efficiency as well as the therapeutic benefit for glucocorticoid loaded anti-MUC5-NP were studied in a murine colitis model. Quantitative adhesion analyses showed that anti-MUC5-conjugated NP exhibited a much higher binding and selectivity to inflamed tissue compared to PNA-, IgG1- and BSA-NP conjugates used as controls. This bioadhesion efficiency was found to be dependent on the ligand density, present at the NP surface. The binding specificity between anti-MUC5 ligand and inflamed tissues was confirmed by fluorescence imaging. Both anti-MUC5-NP and all other glucocorticoid containing formulations led to a significant mitigation of the experimental colitis, as became evident from the substantial reduction of myeloperoxidase activity and pro-inflammatory cytokine concentrations (TNF-α, IL-1ß). Targeted NP by using anti-MUC5 appears to be a very promising tool in future treatment of various types of local disorders affecting the gastro-intestinal tract but not limited to colitis.


Asunto(s)
Colitis , Nanopartículas , Ratones , Animales , Glucocorticoides/uso terapéutico , Ligandos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Nanopartículas/química , Resultado del Tratamiento , Colon/diagnóstico por imagen , Colon/metabolismo
2.
Anal Biochem ; 502: 24-35, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26954998

RESUMEN

G protein-coupled receptors (GPCRs) mediate many important physiological functions and are considered as one of the most successful therapeutic target classes for a wide spectrum of diseases. Drug discovery projects generally benefit from a broad range of experimental approaches for screening compound libraries and for the characterization of binding modes of drug candidates. Owing to the difficulties in solubilizing and purifying GPCRs, assay formats have been so far mainly limited to cell-based functional assays and radioligand binding assays. In this study, we used fluorescence cross-correlation spectroscopy (FCCS) to analyze the interaction of detergent-solubilized receptors to various types of GPCR ligands: endogenous peptides, small molecules, and a large surrogate antagonist represented by a blocking monoclonal antibody. Our work demonstrates the suitability of the homogeneous and time-resolved FCCS assay format for a robust, high-throughput determination of receptor-ligand binding affinities and kinetic rate constants for various therapeutically relevant GPCRs.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Colorantes Fluorescentes/química , Péptidos/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Anticuerpos Monoclonales/química , Sitios de Unión/efectos de los fármacos , Células Cultivadas , Células HEK293 , Humanos , Cinética , Ligandos , Péptidos/química , Receptores Acoplados a Proteínas G/química , Bibliotecas de Moléculas Pequeñas/química , Espectrometría de Fluorescencia , Factores de Tiempo
3.
J Antimicrob Chemother ; 69(4): 1005-16, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24335485

RESUMEN

OBJECTIVES: Artemisinin and artemisinin semi-synthetic derivatives (collectively known as endoperoxides) are first-line antimalarials for the treatment of uncomplicated and severe malaria. Endoperoxides display very fast killing rates and are generally recalcitrant to parasite resistance development. These key pharmacodynamic features are a result of a complex mechanism of action, the details of which lack consensus. Here, we report on the primary physiological events leading to parasite death. METHODS: Parasite mitochondrial (ΔΨm) and plasma membrane (ΔΨp) electrochemical potentials were measured using real-time single-cell imaging following exposure to pharmacologically relevant concentrations of endoperoxides (artemisinin, dihydroartemisinin, artesunate and the synthetic tetraoxane RKA182). In addition, mitochondrial electron transport chain components NADH:quinone oxidoreductase (alternative complex I), bc1 (complex III) and cytochrome oxidase (complex IV) were investigated to determine their functional sensitivity to the various endoperoxides. RESULTS: Parasite exposure to endoperoxides resulted in rapid depolarization of parasite ΔΨm and ΔΨp. The rate of depolarization was decreased in the presence of a reactive oxygen species (ROS) scavenger and Fe(3+) chelators. Depolarization of ΔΨm by endoperoxides is not believed to be through the inhibition of mitochondrial electron transport chain components, owing to the lack of significant inhibition when assayed directly. CONCLUSIONS: The depolarization of ΔΨm and ΔΨp is shown to be mediated via the generation of ROS that are initiated by iron bioactivation of endoperoxides and/or catalysed by iron-dependent oxidative stress. These data are discussed in the context of current hypotheses concerning the mode of action of endoperoxides.


Asunto(s)
Artemisininas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Peroxidasas/farmacología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Especies Reactivas de Oxígeno/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Hierro/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/fisiología , Estrés Oxidativo
4.
Future Med Chem ; 5(13): 1573-91, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24024949

RESUMEN

Despite intense efforts, there has not been a truly new antimalarial, possessing a novel mechanism of action, registered for over 10 years. By virtue of a novel mode of action, it is hoped that the global challenge of multidrug-resistant parasites can be overcome, as well as developing drugs that possess prophylaxis and/or transmission-blocking properties, towards an elimination agenda. Many target-based and whole-cell screening drug development programs have been undertaken in recent years and here an overview of specific projects that have focused on targeting the parasite's mitochondrial electron transport chain is presented. Medicinal chemistry activity has largely focused on inhibitors of the parasite cytochrome bc1 Complex (Complex III) including acridinediones, pyridones and quinolone aryl esters, as well as inhibitors of dihydroorotate dehydrogenase that includes triazolopyrimidines and benzimidazoles. Common barriers to progress and opportunities for novel chemistry and potential additional electron transport chain targets are discussed in the context of the target candidate profiles for uncomplicated malaria.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/enzimología , Animales , Antimaláricos/uso terapéutico , Descubrimiento de Drogas , Transporte de Electrón , Complejo III de Transporte de Electrones/metabolismo , Humanos , Malaria Falciparum/parasitología , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Modelos Moleculares , Terapia Molecular Dirigida , Plasmodium falciparum/efectos de los fármacos
5.
Proc Natl Acad Sci U S A ; 109(21): 8298-303, 2012 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-22566611

RESUMEN

There is an urgent need for new antimalarial drugs with novel mechanisms of action to deliver effective control and eradication programs. Parasite resistance to all existing antimalarial classes, including the artemisinins, has been reported during their clinical use. A failure to generate new antimalarials with novel mechanisms of action that circumvent the current resistance challenges will contribute to a resurgence in the disease which would represent a global health emergency. Here we present a unique generation of quinolone lead antimalarials with a dual mechanism of action against two respiratory enzymes, NADH:ubiquinone oxidoreductase (Plasmodium falciparum NDH2) and cytochrome bc(1). Inhibitor specificity for the two enzymes can be controlled subtly by manipulation of the privileged quinolone core at the 2 or 3 position. Inhibitors display potent (nanomolar) activity against both parasite enzymes and against multidrug-resistant P. falciparum parasites as evidenced by rapid and selective depolarization of the parasite mitochondrial membrane potential, leading to a disruption of pyrimidine metabolism and parasite death. Several analogs also display activity against liver-stage parasites (Plasmodium cynomolgi) as well as transmission-blocking properties. Lead optimized molecules also display potent oral antimalarial activity in the Plasmodium berghei mouse malaria model associated with favorable pharmacokinetic features that are aligned with a single-dose treatment. The ease and low cost of synthesis of these inhibitors fulfill the target product profile for the generation of a potent, safe, and inexpensive drug with the potential for eventual clinical deployment in the control and eradication of falciparum malaria.


Asunto(s)
Antimaláricos/farmacología , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/prevención & control , Plasmodium falciparum/efectos de los fármacos , Piridinas/farmacología , Quinolonas/farmacología , Animales , Antimaláricos/química , Células Cultivadas , Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Hepatocitos/citología , Hepatocitos/parasitología , Macaca mulatta , Malaria Falciparum/parasitología , Masculino , Ratones , Ratones Endogámicos , Mitocondrias/efectos de los fármacos , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/crecimiento & desarrollo , Plasmodium cynomolgi/efectos de los fármacos , Plasmodium cynomolgi/crecimiento & desarrollo , Plasmodium falciparum/crecimiento & desarrollo , Piridinas/química , Quinolonas/química
6.
Antimicrob Agents Chemother ; 56(7): 3739-47, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22547613

RESUMEN

The mitochondrial bc(1) complex is a multisubunit enzyme that catalyzes the transfer of electrons from ubiquinol to cytochrome c coupled to the vectorial translocation of protons across the inner mitochondrial membrane. The complex contains two distinct quinone-binding sites, the quinol oxidation site of the bc(1) complex (Q(o)) and the quinone reduction site (Q(i)), located on opposite sides of the membrane within cytochrome b. Inhibitors of the Q(o) site such as atovaquone, active against the bc(1) complex of Plasmodium falciparum, have been developed and formulated as antimalarial drugs. Unfortunately, single point mutations in the Q(o) site can rapidly render atovaquone ineffective. The development of drugs that could circumvent cross-resistance with atovaquone is needed. Here, we report on the mode of action of a potent inhibitor of P. falciparum proliferation, 1-hydroxy-2-dodecyl-4(1H)quinolone (HDQ). We show that the parasite bc(1) complex--from both control and atovaquone-resistant strains--is inhibited by submicromolar concentrations of HDQ, indicating that the two drugs have different targets within the complex. The binding site of HDQ was then determined by using a yeast model. Introduction of point mutations into the Q(i) site, namely, G33A, H204Y, M221Q, and K228M, markedly decreased HDQ inhibition. In contrast, known inhibitor resistance mutations at the Q(o) site did not cause HDQ resistance. This study, using HDQ as a proof-of-principle inhibitor, indicates that the Q(i) site of the bc(1) complex is a viable target for antimalarial drug development.


Asunto(s)
Antimaláricos/farmacología , Complejo III de Transporte de Electrones/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Quinolonas/farmacología , Antimaláricos/síntesis química , Antimaláricos/química , Sitios de Unión/efectos de los fármacos , Quinolonas/síntesis química , Quinolonas/química
7.
J Biol Chem ; 287(13): 9731-9741, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22282497

RESUMEN

Atovaquone is an anti-malarial drug used in combination with proguanil (e.g. Malarone(TM)) for the curative and prophylactic treatment of malaria. Atovaquone, a 2-hydroxynaphthoquinone, is a competitive inhibitor of the quinol oxidation (Q(o)) site of the mitochondrial cytochrome bc(1) complex. Inhibition of this enzyme results in the collapse of the mitochondrial membrane potential, disruption of pyrimidine biosynthesis, and subsequent parasite death. Resistance to atovaquone in the field is associated with point mutations in the Q(o) pocket of cytochrome b, most notably near the conserved Pro(260)-Glu(261)-Trp(262)-Tyr(263) (PEWY) region in the ef loop). The effect of this mutation has been extensively studied in model organisms but hitherto not in the parasite itself. Here, we have performed a molecular and biochemical characterization of an atovaquone-resistant field isolate, TM902CB. Molecular analysis of this strain reveals the presence of the Y268S mutation in cytochrome b. The Y268S mutation is shown to confer a 270-fold shift of the inhibitory constant (K(i)) for atovaquone with a concomitant reduction in the V(max) of the bc(1) complex of ∼40% and a 3-fold increase in the observed K(m) for decylubiquinol. Western blotting analyses reveal a reduced iron-sulfur protein content in Y268S bc(1) suggestive of a weakened interaction between this subunit and cytochrome b. Gene expression analysis of the TM902CB strain reveals higher levels of expression, compared with the 3D7 (atovaquone-sensitive) control strain in bc(1) and cytochrome c oxidase genes. It is hypothesized that the observed differential expression of these and other key genes offsets the fitness cost resulting from reduced bc(1) activity.


Asunto(s)
Antimaláricos/farmacología , Atovacuona/farmacología , Citocromos b/biosíntesis , Resistencia a Medicamentos , Regulación Enzimológica de la Expresión Génica , Mutación Missense , Plasmodium falciparum/enzimología , Proteínas Protozoarias/biosíntesis , Sustitución de Aminoácidos , Citocromos b/genética , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Plasmodium falciparum/genética , Proguanil/farmacología , Proteínas Protozoarias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...