Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Chem Biol ; 12(9): 709-16, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27398998

RESUMEN

G-protein-coupled receptor (GPCR) ligands function by stabilizing multiple, functionally distinct receptor conformations. This property underlies the ability of 'biased agonists' to activate specific subsets of a given receptor's signaling profile. However, stabilizing distinct active GPCR conformations to enable structural characterization of mechanisms underlying GPCR activation remains difficult. These challenges have accentuated the need for receptor tools that allosterically stabilize and regulate receptor function through unique, previously unappreciated mechanisms. Here, using a highly diverse RNA library combined with advanced selection strategies involving state-of-the-art next-generation sequencing and bioinformatics analyses, we identify RNA aptamers that bind a prototypical GPCR, the ß2-adrenoceptor (ß2AR). Using biochemical, pharmacological, and biophysical approaches, we demonstrate that these aptamers bind with nanomolar affinity at defined surfaces of the receptor, allosterically stabilizing active, inactive, and ligand-specific receptor conformations. The discovery of RNA aptamers as allosteric GPCR modulators significantly expands the diversity of ligands available to study the structural and functional regulation of GPCRs.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Regulación Alostérica/efectos de los fármacos , Aptámeros de Nucleótidos/química , Benzoxazinas/química , Benzoxazinas/farmacología , Humanos , Modelos Moleculares , Conformación Proteica , Receptores Adrenérgicos beta 2/química
2.
J Biol Chem ; 286(31): 27255-65, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21659511

RESUMEN

Aminopeptidases catalyze N-terminal peptide bond hydrolysis and occupy many diverse roles across all domains of life. Here we present evidence that an M1-family aminopeptidase, PfA-M1, has been recruited to specialized roles in the human malaria parasite Plasmodium falciparum. PfA-M1 is abundant in two subcellular compartments in asexual intraerythrocytic parasites; that is, the food vacuole, where the catabolism of host hemoglobin takes place, and the nucleus. A unique N-terminal extension contributes to the observed dual targeting by providing a signal peptide and putative alternate translation initiation sites. PfA-M1 exists as two major isoforms, a nuclear 120-kDa species and a processed species consisting of a complex of 68- and 35-kDa fragments. PfA-M1 is both stable and active at the acidic pH of the food vacuole lumen. Determination of steady-state kinetic parameters for both aminoacyl-ß-naphthylamide and unmodified dipeptide substrates over the pH range 5.0-8.5 reveals that k(cat) is relatively insensitive to pH, whereas K(m) increases at pH values below 6.5. At the pH of the food vacuole lumen (5.0-5.5), the catalytic efficiency of PfA-M1 remains high. Consistent with the kinetic data, the affinity of peptidic competitive inhibitors is diminished at acidic pH. Together, these results support a catalytic role for PfA-M1 in the food vacuole and indicate the importance of evaluating the potency of peptidic inhibitors at physiologically relevant pH values. They also suggest a second, distinct function for this enzyme in the parasite nucleus.


Asunto(s)
Aminopeptidasas/metabolismo , Hemoglobinas/metabolismo , Plasmodium falciparum/enzimología , Vacuolas/metabolismo , Secuencia de Aminoácidos , Aminopeptidasas/química , Aminopeptidasas/aislamiento & purificación , Animales , Secuencia de Bases , Western Blotting , Cartilla de ADN , Eritrocitos/parasitología , Humanos , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
3.
Nanomedicine ; 8(5): 673-81, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21889476

RESUMEN

Control over thrombin activity is much desired to regulate blood clotting in surgical and therapeutic situations. Thrombin-binding RNA and DNA aptamers have been used to inhibit thrombin activity and thus the coagulation cascade. Soluble DNA aptamers, as well as two different aptamers tethered by a flexible single-strand linker, have been shown to possess anticoagulant activity. Here, we link multiple aptamers at programmed positions on DNA nanostructures to optimize spacing and orientation of the aptamers and thereby to maximize anticoagulant activity in functional assays. By judicious engineering of the DNA nanostructures, we have created a novel, functional DNA nanostructure, which is a multi-aptamer inhibitor with activity eightfold higher than free aptamer. Reversal of the thrombin inhibition was also achieved by the use of single-stranded DNA antidotes, thus enabling significant control over blood coagulation. FROM THE CLINICAL EDITOR: Thrombin inhibition via DNA aptamers has recently become a possibility. In this study, thrombin-binding DNA aptamers were further optimized by nanoscale organization on DNA nanostructures. The authors have created a novel, functional DNA nanostructure, which is a multi-aptamer inhibitor with activity eightfold higher than that of free aptamer. Reversal of thrombin inhibition was also achieved by single-stranded DNA antidotes, enabling significant control over the coagulation pathway.


Asunto(s)
Anticoagulantes/farmacología , Aptámeros de Nucleótidos/farmacología , ADN/química , Trombina/química , Anticoagulantes/química , Aptámeros de Nucleótidos/química , Coagulación Sanguínea/efectos de los fármacos , Humanos , Nanoestructuras/química , Plasma/química , Trombina/antagonistas & inhibidores
4.
RNA ; 15(12): 2105-11, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19846574

RESUMEN

Thrombin is a multifunctional protease that plays a key role in hemostasis, thrombosis, and inflammation. Most thrombin inhibitors currently used as antithrombotic agents target thrombin's active site and inhibit all of its myriad of activities. Exosites 1 and 2 are distinct regions on the surface of thrombin that provide specificity to its proteolytic activity by mediating binding to substrates, receptors, and cofactors. Exosite 1 mediates binding and cleavage of fibrinogen, proteolytically activated receptors, and some coagulation factors, while exosite 2 mediates binding to heparin and to platelet receptor GPIb-IX-V. The crystal structures of two nucleic acid ligands bound to thrombin have been solved. Previously Padmanabhan and colleagues solved the structure of a DNA aptamer bound to exosite 1 and we reported the structure of an RNA aptamer bound to exosite 2 on thrombin. Based upon these structural studies we speculated that the two aptamers would not compete for binding to thrombin. We observe that simultaneously blocking both exosites with the aptamers leads to synergistic inhibition of thrombin-dependent platelet activation and procoagulant activity. This combination of exosite 1 and exosite 2 inhibitors may provide a particularly effective antithrombotic approach.


Asunto(s)
Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Fibrinolíticos/química , Fibrinolíticos/farmacología , Trombina/química , Trombina/metabolismo , Aptámeros de Nucleótidos/metabolismo , Sinergismo Farmacológico , Activación Enzimática , Fibrinolíticos/metabolismo , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , Selectina-P/metabolismo , Activación Plaquetaria , Unión Proteica , Estructura Terciaria de Proteína , Trombina/antagonistas & inhibidores
5.
J Am Chem Soc ; 132(41): 14481-6, 2010 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-20863133

RESUMEN

Architectural designs for DNA nanostructures typically fall within one of two broad categories: tile-based designs (assembled from chemically synthesized oligonucleotides) and origami designs (woven structures employing a biological scaffold strand and synthetic staple strands). Both previous designs typically contain many Holliday-type multi-arm junctions. Here we describe the design, implementation, and testing of a unique architectural strategy incorporating some aspects of each of the two previous design categories but without multi-arm junction motifs. Goals for the new design were to use only chemically synthesized DNA, to minimize the number of component strands, and to mimic the back-and-forth, woven strand routing of the origami architectures. The resulting architectural strategy employs "weave tiles" formed from only two oligonucleotides as basic building blocks, thus decreasing the burden of matching multiple strand stoichiometries compared to previous tile-based architectures and resulting in a structurally flexible tile. As an example application, we have shown that the four-helix weave tile can be used to increase the anticoagulant activity of thrombin-binding aptamers in vitro.


Asunto(s)
ADN/química , Nanotecnología , Secuencia de Bases , Microscopía de Fuerza Atómica , Datos de Secuencia Molecular
6.
Nat Biotechnol ; 36(7): 606-613, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29863725

RESUMEN

Unfractionated heparin (UFH), the standard anticoagulant for cardiopulmonary bypass (CPB) surgery, carries a risk of post-operative bleeding and is potentially harmful in patients with heparin-induced thrombocytopenia-associated antibodies. To improve the activity of an alternative anticoagulant, the RNA aptamer 11F7t, we solved X-ray crystal structures of the aptamer bound to factor Xa (FXa). The finding that 11F7t did not bind the catalytic site suggested that it could complement small-molecule FXa inhibitors. We demonstrate that combinations of 11F7t and catalytic-site FXa inhibitors enhance anticoagulation in purified reaction mixtures and plasma. Aptamer-drug combinations prevented clot formation as effectively as UFH in human blood circulated in an extracorporeal oxygenator circuit that mimicked CPB, while avoiding side effects of UFH. An antidote could promptly neutralize the anticoagulant effects of both FXa inhibitors. Our results suggest that drugs and aptamers with shared targets can be combined to exert more specific and potent effects than either agent alone.


Asunto(s)
Anticoagulantes/administración & dosificación , Inhibidores del Factor Xa/administración & dosificación , Factor Xa/química , Hemorragia Posoperatoria/tratamiento farmacológico , Anticoagulantes/química , Aptámeros de Nucleótidos/administración & dosificación , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/genética , Puente Cardiopulmonar/efectos adversos , Cristalografía por Rayos X , Combinación de Medicamentos , Factor Xa/genética , Inhibidores del Factor Xa/química , Heparina/efectos adversos , Humanos , Hemorragia Posoperatoria/genética , Hemorragia Posoperatoria/patología , Conformación Proteica/efectos de los fármacos
7.
Nucleic Acid Ther ; 26(1): 1-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26584417

RESUMEN

Potent and rapid-onset anticoagulation is required for several clinical settings, including cardiopulmonary bypass surgery. In addition, because anticoagulation is associated with increased bleeding following surgery, the ability to rapidly reverse such robust anticoagulation is also important. Previously, we observed that no single aptamer was as potent as heparin for anticoagulating blood. However, we discovered that combinations of two aptamers were as potent as heparin. Herein, we sought to combine two individual anticoagulant aptamers into a single bivalent RNA molecule in an effort to generate a single molecule that retained the potent anticoagulant activity of the combination of individual aptamers. We created four bivalent aptamers that can inhibit Factor X/Xa and prothrombin/thrombin and anticoagulate plasma, as well as the combination of individual aptamers. Detailed characterization of the shortest bivalent aptamer indicates that each aptamer retains full binding and functional activity when presented in the bivalent context. Finally, reversal of this bivalent aptamer with a single antidote was explored, and anticoagulant activity could be rapidly turned off in a dose-dependent manner. These studies demonstrate that bivalent anticoagulant aptamers represent a novel and potent approach to actively and reversibly control coagulation.


Asunto(s)
Anticoagulantes/farmacología , Antídotos/farmacología , Aptámeros de Nucleótidos/metabolismo , Factores de Coagulación Sanguínea/metabolismo , Serina Proteasas/metabolismo , Aptámeros de Nucleótidos/química , Secuencia de Bases , Factores de Coagulación Sanguínea/genética , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Serina Proteasas/genética
8.
Metallomics ; 8(9): 951-62, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27157188

RESUMEN

The development of resistance to cisplatin (cDDP) is commonly accompanied by reduced drug uptake or increased efflux. Previous studies in yeast and murine embryonic fibroblasts have reported that the copper (Cu) transporters and chaperones participate in the uptake, efflux, and intracellular distribution of cDDP. However, there is conflicting data from studies in human cells. We used CRISPR-Cas9 genome editing to individually knock out the human copper transporters CTR1 and CTR2 and the copper chaperones ATOX1 and CCS. Isogenic knockout cell lines were generated in both human HEK-293T and ovarian carcinoma OVCAR8 cells. All knockout cell lines had slowed growth compared to parental cells, small changes in basal Cu levels, and varying sensitivities to Cu depending on the gene targeted. However, all of the knockouts demonstrated only modest 2 to 5-fold changes in cDDP sensitivity that did not differ from the range of sensitivities of 10 wild type clones grown from the same parental cell population. We conclude that, under basal conditions, loss of CTR1, CTR2, ATOX1, or CCS does not produce a change in cisplatin sensitivity that exceeds the variance found within the parental population, suggesting that they are not essential to the mechanism by which cDDP enters these cell lines and is transported to the nucleus.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cisplatino/farmacología , Cobre/metabolismo , Resistencia a Antineoplásicos , Chaperonas Moleculares/metabolismo , Neoplasias Ováricas/patología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas Transportadoras de Cobre , Transportador de Cobre 1 , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Proteínas SLC31 , Ensayos Antitumor por Modelo de Xenoinjerto
9.
ACS Comb Sci ; 18(8): 461-74, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27314875

RESUMEN

Botulism is caused by potent and specific bacterial neurotoxins that infect host neurons and block neurotransmitter release. Treatment for botulism is limited to administration of an antitoxin within a short time window, before the toxin enters neurons. Alternatively, current botulism drug development targets the toxin light chain, which is a zinc-dependent metalloprotease that is delivered into neurons and mediates long-term pathology. Several groups have identified inhibitory small molecules, peptides, or aptamers, although no molecule has advanced to the clinic due to a lack of efficacy in advanced models. Here we used a homogeneous high-throughput enzyme assay to screen three libraries of drug-like small molecules for new chemotypes that modulate recombinant botulinum neurotoxin light chain activity. High-throughput screening of 97088 compounds identified numerous small molecules that activate or inhibit metalloprotease activity. We describe four major classes of inhibitory compounds identified, detail their structure-activity relationships, and assess their relative inhibitory potency. A previously unreported chemotype in any context of enzyme inhibition is described with potent submicromolar inhibition (Ki = 200-300 nM). Additional detailed kinetic analyses and cellular cytotoxicity assays indicate the best compound from this series is a competitive inhibitor with cytotoxicity values around 4-5 µM. Given the potency and drug-like character of these lead compounds, further studies, including cellular activity assays and DMPK analysis, are justified.


Asunto(s)
Toxinas Botulínicas/antagonistas & inhibidores , Inhibidores de Proteasas/química , Bibliotecas de Moléculas Pequeñas/química , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Ratones , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/toxicidad , Pirazoles/química , Piridinas/química , Quinolinas/química , Relación Estructura-Actividad , Tiadiazoles/química
10.
Curr Top Med Chem ; 14(18): 2062-80, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25335886

RESUMEN

Botulinum neurotoxins (BoNTs) are a class of bacterial neurotoxins that are the most potent toxic compounds reported to date. Exposure to relatively low concentrations of the toxin protein can result in major muscle paralysis, which may result in death in severe cases. In addition to their role in natural human disease, BoNTs are currently under close scrutiny because of their potential to be used as biowarfare agents. Clinical treatment options for botulism are currently limited, and finite stockpiles of antitoxin exist. In light of current bioterrorist threats, researchers have focused on identifying new molecules that can be applied to either sensitive toxin detection or improved clinical treatment. High-throughput screening (HTS) is a laboratory technique commonly employed to screen large libraries of diverse compounds based on specific compound binding capabilities or function. Here we review existing HTS platforms that have been applied to identify novel BoNT diagnostic or therapeutic agents. HTS platforms for screening antibodies, peptides, small molecules, and aptamers are described, as well as the screening results and current progress of the identified compounds.


Asunto(s)
Antídotos/farmacología , Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Botulismo/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento/métodos , Peptidomiméticos/farmacología , Animales , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/farmacología , Antídotos/química , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Bioterrorismo/prevención & control , Toxinas Botulínicas Tipo A/química , Toxinas Botulínicas Tipo A/toxicidad , Botulismo/patología , Quelantes/química , Quelantes/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Biblioteca de Péptidos , Peptidomiméticos/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Transmisión Sináptica/efectos de los fármacos
11.
Chem Biol ; 21(8): 935-44, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25065530

RESUMEN

Coordinated enzymatic reactions regulate blood clot generation. To explore the contributions of various coagulation enzymes in this process, we utilized a panel of aptamers against factors VIIa, IXa, Xa, and prothrombin. Each aptamer dose-dependently inhibited clot formation, yet none was able to completely impede this process in highly procoagulant settings. However, several combinations of two aptamers synergistically impaired clot formation. One extremely potent aptamer combination was able to maintain human blood fluidity even during extracorporeal circulation, a highly procoagulant setting encountered during cardiopulmonary bypass surgery. Moreover, this aptamer cocktail could be rapidly reversed with antidotes to restore normal hemostasis, indicating that even highly potent aptamer combinations can be rapidly controlled. These studies highlight the potential utility of using sets of aptamers to probe the functions of proteins in molecular pathways for research and therapeutic ends.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Factores de Coagulación Sanguínea/antagonistas & inhibidores , Coagulación Sanguínea/efectos de los fármacos , Protrombina/antagonistas & inhibidores , Factores de Coagulación Sanguínea/metabolismo , Pruebas de Coagulación Sanguínea , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Voluntarios Sanos , Humanos , Protrombina/metabolismo , Relación Estructura-Actividad
12.
J Med Chem ; 57(3): 669-76, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24387280

RESUMEN

Botulinum neurotoxins (BoNT) are the most potent toxins known and a significant bioterrorist threat. Few small molecule compounds have been identified that are active in cell-based or animal models, potentially due to toxin enzyme plasticity. Here we screened commercially available quinolinols, as well as synthesized hydroxyquinolines. Seventy-two compounds had IC50 values below 10 µM, with the best compound exhibiting submicromolar inhibition (IC50 = 0.8 µM). Structure-activity relationship trends showed that the enzyme tolerates various substitutions at R1 but has a clear preference for bulky aryl amide groups at R2, while methylation at R3 increased inhibitor potency. Evaluation of the most potent compounds in an ADME panel showed that these compounds possess poor solubility at pH 6.8, but display excellent solubility at low pH, suggesting that oral dosing may be possible. Our data show the potential of quinolinol compounds as BoNT therapeutics due to their good in vitro potencies and favorable ADME properties.


Asunto(s)
Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Hidroxiquinolinas/química , Animales , Dominio Catalítico , Humanos , Concentración de Iones de Hidrógeno , Hidroxiquinolinas/síntesis química , Hidroxiquinolinas/farmacología , Técnicas In Vitro , Microsomas Hepáticos/metabolismo , Ratas , Ratas Sprague-Dawley , Solubilidad , Relación Estructura-Actividad
13.
J Vis Exp ; (82): 50908, 2013 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-24430674

RESUMEN

Botulinum neurotoxin (BoNT) is a potent and potentially lethal bacterial toxin that binds to host motor neurons, is internalized into the cell, and cleaves intracellular proteins that are essential for neurotransmitter release. BoNT is comprised of a heavy chain (HC), which mediates host cell binding and internalization, and a light chain (LC), which cleaves intracellular host proteins essential for acetylcholine release. While therapies that inhibit toxin binding/internalization have a small time window of administration, compounds that target intracellular LC activity have a much larger time window of administrations, particularly relevant given the extremely long half-life of the toxin. In recent years, small molecules have been heavily analyzed as potential LC inhibitors based on their increased cellular permeability relative to larger therapeutics (peptides, aptamers, etc.). Lead identification often involves high-throughput screening (HTS), where large libraries of small molecules are screened based on their ability to modulate therapeutic target function. Here we describe a FRET-based assay with a commercial BoNT/A LC substrate and recombinant LC that can be automated for HTS of potential BoNT inhibitors. Moreover, we describe a manual technique that can be used for follow-up secondary screening, or for comparing the potency of several candidate compounds.


Asunto(s)
Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Toxinas Botulínicas Tipo A/química , Toxinas Botulínicas Tipo A/inmunología , Proteína 25 Asociada a Sinaptosomas/metabolismo
14.
Nat Med ; 15(10): 1224-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19801990

RESUMEN

With an ever increasing number of people taking numerous medications, the need to safely administer drugs and limit unintended side effects has never been greater. Antidote control remains the most direct means to counteract acute side effects of drugs, but, unfortunately, it has been challenging and cost prohibitive to generate antidotes for most therapeutic agents. Here we describe the development of a set of antidote molecules that are capable of counteracting the effects of an entire class of therapeutic agents based upon aptamers. These universal antidotes exploit the fact that, when systemically administered, aptamers are the only free extracellular oligonucleotides found in circulation. We show that protein- and polymer-based molecules that capture oligonucleotides can reverse the activity of several aptamers in vitro and counteract aptamer activity in vivo. The availability of universal antidotes to control the activity of any aptamer suggests that aptamers may be a particularly safe class of therapeutics.


Asunto(s)
Anticoagulantes/efectos adversos , Antídotos/farmacología , Aptámeros de Nucleótidos/farmacología , Oligonucleótidos/farmacología , Anticoagulantes/farmacología , Antídotos/administración & dosificación , Aptámeros de Nucleótidos/clasificación , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Factor IX/antagonistas & inhibidores , Inhibidores del Factor Xa , Humanos , Conformación de Ácido Nucleico/efectos de los fármacos , Protaminas/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA