Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Bioorg Med Chem ; 28(23): 115762, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32992249

RESUMEN

Human factor XIa (hFXIa) has emerged as an attractive target for development of new anticoagulants that promise higher level of safety. Different strategies have been adopted so far for the design of anti-hFXIa molecules including competitive and non-competitive inhibition. Of these, allosteric dysfunction of hFXIa's active site is especially promising because of the possibility of controlled reduction in activity that may offer a route to safer anticoagulants. In this work, we assess fragment-based design approach to realize a group of novel allosteric hFXIa inhibitors. Starting with our earlier discovery that sulfated quinazolinone (QAO) bind in the heparin-binding site of hFXIa, we developed a group of two dozen dimeric sulfated QAOs with intervening linkers that displayed a progressive variation in inhibition potency. In direct opposition to the traditional wisdom, increasing linker flexibility led to higher potency, which could be explained by computational studies. Sulfated QAO 19S was identified as the most potent and selective inhibitor of hFXIa. Enzyme inhibition studies revealed that 19S utilizes a non-competitive mechanism of action, which was supported by fluorescence studies showing a classic sigmoidal binding profile. Studies with selected mutants of hFXIa indicated that sulfated QAOs bind in heparin-binding site of the catalytic domain of hFXIa. Overall, the approach of fragment-based design offers considerable promise for designing heparin-binding site-directed allosteric inhibitors of hFXIa.


Asunto(s)
Diseño de Fármacos , Factor XIa/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/química , Regulación Alostérica/efectos de los fármacos , Sitios de Unión , Dominio Catalítico , Dimerización , Factor XIa/metabolismo , Humanos , Cinética , Simulación del Acoplamiento Molecular , Quinazolinonas/química , Quinazolinonas/metabolismo , Quinazolinonas/farmacología , Inhibidores de Serina Proteinasa/metabolismo , Relación Estructura-Actividad , Sulfatos/química
2.
Bioorg Med Chem Lett ; 28(6): 1101-1105, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29459207

RESUMEN

Despite the development of promising direct oral anticoagulants, which are all orthosteric inhibitors, a sizable number of patients suffer from bleeding complications. We have hypothesized that allosterism based on the heparin-binding exosites presents a major opportunity to induce sub-maximal inhibition of coagulation proteases, thereby avoiding/reducing bleeding risk. We present the design of a group of sulfated benzofuran dimers that display heparin-binding site-dependent partial allosteric inhibition of thrombin against fibrinogen (ΔY = 55-75%), the first time that a small molecule (MW  < 800) has been found to thwart macromolecular cleavage by a monomeric protease in a controlled manner. The work leads to the promising concept that it should be possible to develop allosteric inhibitors that reduce clotting, but do not completely eliminate it, thereby avoiding major bleeding complications that beset anticoagulants today.


Asunto(s)
Benzofuranos/farmacología , Inhibidores de Serina Proteinasa/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfatos/farmacología , Trombina/antagonistas & inhibidores , Benzofuranos/síntesis química , Benzofuranos/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Serina Proteinasa/síntesis química , Inhibidores de Serina Proteinasa/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Sulfatos/química , Trombina/metabolismo
3.
Molecules ; 20(1): 608-24, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25569517

RESUMEN

Plasmin, a key serine protease, plays a major role in clot lysis and extracellular matrix remodeling. Heparin, a natural polydisperse sulfated glycosaminoglycan, is known to allosterically modulate plasmin activity. No small allosteric inhibitor of plasmin has been discovered to date. We screened an in-house library of 55 sulfated, small glycosaminoglycan mimetics based on nine distinct scaffolds and varying number and positions of sulfate groups to discover several promising hits. Of these, a pentasulfated flavonoid-quinazolinone dimer 32 was found to be the most potent sulfated small inhibitor of plasmin (IC50 = 45 µM, efficacy = 100%). Michaelis-Menten kinetic studies revealed an allosteric inhibition of plasmin by these inhibitors. Studies also indicated that the most potent inhibitors are selective for plasmin over thrombin and factor Xa, two serine proteases in coagulation cascade. Interestingly, different inhibitors exhibited different levels of efficacy (40%-100%), an observation alluding to the unique advantage offered by an allosteric process. Overall, our work presents the first small, synthetic allosteric plasmin inhibitors for further rational design.


Asunto(s)
Fibrinolisina/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Sulfatos/metabolismo , Regulación Alostérica/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Factor Xa/metabolismo , Fibrinolisina/antagonistas & inhibidores , Humanos , Hidrólisis/efectos de los fármacos , Cinética , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Sulfatos/química
4.
Bioorg Med Chem Lett ; 22(13): 4467-70, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22627041

RESUMEN

Tumor-associated angiogenesis is a complex process that involves the interplay among several molecular players such as cell-surface heparan sulfate proteoglycans, vascular endothelial growth factors and their cognate receptors. PI-88, a highly sulfonated oligosaccharide, has been shown to have potent anti-angiogenic activity and is currently in clinical trials. However, one of the major drawbacks of large oligosaccharides such as PI-88 is that their synthesis often requires numerous complex synthetic steps. In this study, several novel polysulfonated small molecule carbohydrate mimetics, which can easily be synthesized in fewer steps, are identified as promising inhibitors of angiogenesis in an in vitro tube formation assay.


Asunto(s)
Inhibidores de la Angiogénesis/química , Materiales Biomiméticos/química , Bibliotecas de Moléculas Pequeñas/química , Azufre/química , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/farmacología , Animales , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/farmacología , Bovinos , Evaluación Preclínica de Medicamentos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Oligosacáridos/farmacología , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/farmacología
5.
PLoS One ; 11(7): e0160189, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27467511

RESUMEN

Factor XIIIa (FXIIIa) is a transglutaminase that catalyzes the last step in the coagulation process. Orthostery is the only approach that has been exploited to design FXIIIa inhibitors. Yet, allosteric inhibition of FXIIIa is a paradigm that may offer a key advantage of controlled inhibition over orthosteric inhibition. Such an approach is likely to lead to novel FXIIIa inhibitors that do not carry bleeding risks. We reasoned that targeting a collection of basic amino acid residues distant from FXIIIa's active site by using sulfated glycosaminoglycans (GAGs) or non-saccharide GAG mimetics (NSGMs) would lead to the discovery of the first allosteric FXIIIa inhibitors. We tested a library of 22 variably sulfated GAGs and NSGMs against human FXIIIa to discover promising hits. Interestingly, although some GAGs bound to FXIIIa better than NSGMs, no GAG displayed any inhibition. An undecasulfated quercetin analog was found to inhibit FXIIIa with reasonable potency (efficacy of 98%). Michaelis-Menten kinetic studies revealed an allosteric mechanism of inhibition. Fluorescence studies confirmed close correspondence between binding affinity and inhibition potency, as expected for an allosteric process. The inhibitor was reversible and at least 9-fold- and 26-fold selective over two GAG-binding proteins factor Xa (efficacy of 71%) and thrombin, respectively, and at least 27-fold selective over a cysteine protease papain. The inhibitor also inhibited the FXIIIa-mediated polymerization of fibrin in vitro. Overall, our work presents the proof-of-principle that FXIIIa can be allosterically modulated by sulfated non-saccharide agents much smaller than GAGs, which should enable the design of selective and safe anticoagulants.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Factor XIIIa/antagonistas & inhibidores , Glicosaminoglicanos/farmacología , Imitación Molecular , Regulación Alostérica , Inhibidores Enzimáticos/química , Fibrina/química , Glicosaminoglicanos/química , Cinética , Polimerizacion , Relación Estructura-Actividad
6.
Methods Mol Biol ; 1229: 49-67, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25325944

RESUMEN

In nearly all cases of biological activity of sulfated GAGs, the sulfate group(s) are critical for interacting with target proteins. A growing paradigm is that appropriate small, sulfated, nonsaccharide GAG mimetics can be designed to either mimic or interfere with the biological functions of natural GAG sequences resulting in the discovery of either antagonist or agonist agents. A number of times these sulfated NSGMs can be computationally designed based on the parent GAG-protein interaction. The small sulfated NSGMs may possess considerable aromatic character so as to engineer hydrophobic, hydrogen-bonding, Coulombic or cation-pi forces in their interactions with target protein(s) resulting in higher specificity of action relative to parent GAGs. The sulfated NSGMs can be easily synthesized in one step from appropriate natural polyphenols through chemical sulfation under microwave-based conditions. We describe step-by-step procedures to perform microwave-based sulfation of several small polyphenol scaffolds so as to prepare homogenous NSGMs containing one to more than 10 sulfate groups per molecule in high yields.


Asunto(s)
Bioquímica/métodos , Materiales Biomiméticos/síntesis química , Glicosaminoglicanos/síntesis química , Polifenoles/química , Sulfatos/química , Cromatografía Líquida de Alta Presión , Glicosaminoglicanos/química , Interacciones Hidrofóbicas e Hidrofílicas
7.
ACS Chem Biol ; 9(8): 1826-33, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24968014

RESUMEN

Selective targeting of cancer stem-like cells (CSCs) is a paradigm-shifting approach. We hypothesized that CSCs can be targeted by interfering with functions of sulfated glycosaminoglycans, which play key roles in cancer cell growth, invasion and metastasis. We developed a tandem, dual screen strategy involving (1) assessing inhibition of monolayer versus spheroid growth and (2) assessing inhibition of primary versus secondary spheroid growth to identify G2.2, a unique sulfated nonsaccharide GAG mimetic (NSGM) from a focused library of 53 molecules, as a selective inhibitor of colon CSCs. The NSGM down-regulated several CSC markers through regulation of gene transcription, while closely related, inactive NSGMs G1.4 and G4.1 demonstrated no such changes. G2.2's effects on CSCs were mediated, in part, through induction of apoptosis and inhibition of self-renewal factors. Overall, this work presents the proof-of-principle that CSCs can be selectively targeted through novel NSGMs, which are likely to advance fundamental understanding on CSCs while also aiding development of novel therapeutic agents.


Asunto(s)
Neoplasias del Colon/patología , Glicosaminoglicanos/farmacología , Imitación Molecular , Células Madre Neoplásicas/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Células Madre Neoplásicas/patología
8.
J Med Chem ; 56(6): 2415-28, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23451707

RESUMEN

To discover promising sulfated allosteric modulators (SAMs) of glycosaminoglycan-binding proteins (GBPs), such as human factor XIa (FXIa), we screened a library of 26 synthetic, sulfated quinazolin-4(3H)-ones (QAOs) resulting in the identification of six molecules that reduced the Vmax of substrate hydrolysis without influencing the KM. Mutagenesis of residues of the heparin-binding site (HBS) of FXIa introduced a nearly 5-fold loss in inhibition potency supporting recognition of an allosteric site. Fluorescence studies showed a sigmoidal binding profile indicating highly cooperative binding. Competition with a positively charged, heparin-binding polymer did not fully nullify inhibition suggesting importance of hydrophobic forces to binding. This discovery suggests the operation of a dual-element recognition process, which relies on an initial Coulombic attraction of anionic SAMs to the cationic HBS of FXIa that forms a locked complex through tight interaction with an adjacent hydrophobic patch. The dual-element strategy may be widely applicable for discovering SAMs of other GBPs.


Asunto(s)
Dominio Catalítico , Descubrimiento de Drogas , Factor XIa/química , Heparina/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Quinazolinonas/química , Quinazolinonas/farmacología , Regulación Alostérica/efectos de los fármacos , Materiales Biomiméticos/farmacología , Factor XIa/antagonistas & inhibidores , Factor XIa/metabolismo , Glicosaminoglicanos/química , Humanos , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Sulfatos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA