Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(25): e2218896120, 2023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-37327313

RESUMEN

Programmed ferroptotic death eliminates cells in all major organs and tissues with imbalanced redox metabolism due to overwhelming iron-catalyzed lipid peroxidation under insufficient control by thiols (Glutathione (GSH)). Ferroptosis has been associated with the pathogenesis of major chronic degenerative diseases and acute injuries of the brain, cardiovascular system, liver, kidneys, and other organs, and its manipulation offers a promising new strategy for anticancer therapy. This explains the high interest in designing new small-molecule-specific inhibitors against ferroptosis. Given the role of 15-lipoxygenase (15LOX) association with phosphatidylethanolamine (PE)-binding protein 1 (PEBP1) in initiating ferroptosis-specific peroxidation of polyunsaturated PE, we propose a strategy of discovering antiferroptotic agents as inhibitors of the 15LOX/PEBP1 catalytic complex rather than 15LOX alone. Here we designed, synthesized, and tested a customized library of 26 compounds using biochemical, molecular, and cell biology models along with redox lipidomic and computational analyses. We selected two lead compounds, FerroLOXIN-1 and 2, which effectively suppressed ferroptosis in vitro and in vivo without affecting the biosynthesis of pro-/anti-inflammatory lipid mediators in vivo. The effectiveness of these lead compounds is not due to radical scavenging or iron-chelation but results from their specific mechanisms of interaction with the 15LOX-2/PEBP1 complex, which either alters the binding pose of the substrate [eicosatetraenoyl-PE (ETE-PE)] in a nonproductive way or blocks the predominant oxygen channel thus preventing the catalysis of ETE-PE peroxidation. Our successful strategy may be adapted to the design of additional chemical libraries to reveal new ferroptosis-targeting therapeutic modalities.


Asunto(s)
Ferroptosis , Proteínas de Unión a Fosfatidiletanolamina , Glutatión/metabolismo , Hierro/metabolismo , Peroxidación de Lípido , Lípidos , Oxidación-Reducción , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores
2.
Arterioscler Thromb Vasc Biol ; 37(10): 1828-1839, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28775075

RESUMEN

OBJECTIVE: Adequate platelet reactivity is required for maintaining hemostasis. However, excessive platelet reactivity can also lead to the formation of occlusive thrombi. Platelet 12(S)-lipoxygenase (12-LOX), an oxygenase highly expressed in the platelet, has been demonstrated to regulate platelet function and thrombosis ex vivo, supporting a key role for 12-LOX in the regulation of in vivo thrombosis. However, the ability to pharmacologically target 12-LOX in vivo has not been established to date. Here, we studied the effect of the first highly selective 12-LOX inhibitor, ML355, on in vivo thrombosis and hemostasis. APPROACH AND RESULTS: ML355 dose-dependently inhibited human platelet aggregation and 12-LOX oxylipin production, as confirmed by mass spectrometry. Interestingly, the antiplatelet effects of ML355 were reversed after exposure to high concentrations of thrombin in vitro. Ex vivo flow chamber assays confirmed that human platelet adhesion and thrombus formation at arterial shear over collagen were attenuated in whole blood treated with ML355 comparable to aspirin. Oral administration of ML355 in mice showed reasonable plasma drug levels by pharmacokinetic assessment. ML355 treatment impaired thrombus growth and vessel occlusion in FeCl3-induced mesenteric and laser-induced cremaster arteriole thrombosis models in mice. Importantly, hemostatic plug formation and bleeding after treatment with ML355 was minimal in mice in response to laser ablation on the saphenous vein or in a cremaster microvasculature laser-induced rupture model. CONCLUSIONS: Our data strongly support 12-LOX as a key determinant of platelet reactivity in vivo, and inhibition of platelet 12-LOX with ML355 may represent a new class of antiplatelet therapy.


Asunto(s)
Hemostasis/efectos de los fármacos , Inhibidores de la Lipooxigenasa/farmacología , Agregación Plaquetaria/efectos de los fármacos , Sulfonamidas/farmacología , Trombosis/prevención & control , Animales , Relación Dosis-Respuesta a Droga , Humanos , Inhibidores de la Lipooxigenasa/administración & dosificación , Inhibidores de la Lipooxigenasa/sangre , Ratones , Adhesividad Plaquetaria/efectos de los fármacos , Sulfonamidas/administración & dosificación , Sulfonamidas/sangre , Trombina/fisiología
3.
Blood ; 124(14): 2271-9, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25100742

RESUMEN

Platelets are essential in maintaining hemostasis following inflammation or injury to the vasculature. Dysregulated platelet activity often results in thrombotic complications leading to myocardial infarction and stroke. Activation of the FcγRIIa receptor leads to immune-mediated thrombosis, which is often life threatening in patients undergoing heparin-induced thrombocytopenia or sepsis. Inhibiting FcγRIIa-mediated activation in platelets has been shown to limit thrombosis and is the principal target for prevention of immune-mediated platelet activation. In this study, we show for the first time that platelet 12(S)-lipoxygenase (12-LOX), a highly expressed oxylipin-producing enzyme in the human platelet, is an essential component of FcγRIIa-mediated thrombosis. Pharmacologic inhibition of 12-LOX in human platelets resulted in significant attenuation of FcγRIIa-mediated aggregation. Platelet 12-LOX was shown to be essential for FcγRIIa-induced phospholipase Cγ2 activity leading to activation of calcium mobilization, Rap1 and protein kinase C activation, and subsequent activation of the integrin αIIbß3. Additionally, platelets from transgenic mice expressing human FcγRIIa but deficient in platelet 12-LOX, failed to form normal platelet aggregates and exhibited deficiencies in Rap1 and αIIbß3 activation. These results support an essential role for 12-LOX in regulating FcγRIIa-mediated platelet function and identifies 12-LOX as a potential therapeutic target to limit immune-mediated thrombosis.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Plaquetas/metabolismo , Receptores de IgG/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/química , Animales , Calcio/metabolismo , Activación Enzimática , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfolipasa C gamma/metabolismo , Fosforilación , Activación Plaquetaria , Agregación Plaquetaria , Proteína Quinasa C/metabolismo , Transducción de Señal , Trombosis/inmunología
4.
Nat Chem Biol ; 10(4): 298-304, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24531842

RESUMEN

Protein ubiquitination and deubiquitination are central to the control of a large number of cellular pathways and signaling networks in eukaryotes. Although the essential roles of ubiquitination have been established in the eukaryotic DNA damage response, the deubiquitination process remains poorly defined. Chemical probes that perturb the activity of deubiquitinases (DUBs) are needed to characterize the cellular function of deubiquitination. Here we report ML323 (2), a highly potent inhibitor of the USP1-UAF1 deubiquitinase complex with excellent selectivity against human DUBs, deSUMOylase, deneddylase and unrelated proteases. Using ML323, we interrogated deubiquitination in the cellular response to UV- and cisplatin-induced DNA damage and revealed new insights into the requirement of deubiquitination in the DNA translesion synthesis and Fanconi anemia pathways. Moreover, ML323 potentiates cisplatin cytotoxicity in non-small cell lung cancer and osteosarcoma cells. Our findings point to USP1-UAF1 as a key regulator of the DNA damage response and a target for overcoming resistance to the platinum-based anticancer drugs.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Proteínas de Arabidopsis/antagonistas & inhibidores , Daño del ADN/fisiología , Proteínas Nucleares/antagonistas & inhibidores , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Ubiquitinación/efectos de los fármacos , Algoritmos , Butiratos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Ensayo de Unidades Formadoras de Colonias , Daño del ADN/genética , ADN de Neoplasias/antagonistas & inhibidores , ADN de Neoplasias/biosíntesis , Resistencia a Antineoplásicos , Electroforesis en Gel de Poliacrilamida , Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Humanos , Indicadores y Reactivos , Compuestos de Fenilurea/farmacología , Pimozida/farmacología , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/química , Recombinación Genética/efectos de los fármacos , Intercambio de Cromátides Hermanas/efectos de los fármacos
5.
Bioorg Med Chem Lett ; 22(16): 5326-9, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22795331

RESUMEN

Inhibitors of ketohexokinase (KHK) have potential for the treatment of diabetes and obesity. We have continued studies on a pyrimidinopyrimidine series of potent KHK inhibitors by exploring the 2-position substituent (R(3)) that interacts with Asp-27B in the ATP-binding region of KHK (viz. 1, 2; Table 1). We found that increased spacing between the terminal ammonium group and the heterocyclic scaffold (viz. 16-20), such that interaction with Asp-27B is not possible, still results in potent KHK inhibition (IC(50)=15-50 nM). We propose a new interaction with Asp-194, which serves to expand the pyrimidinopyrimidine pharmacophore.


Asunto(s)
Inhibidores Enzimáticos/química , Fructoquinasas/antagonistas & inhibidores , Ligandos , Pirimidinas/química , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Fructoquinasas/metabolismo , Células Hep G2 , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Pirimidinas/síntesis química , Pirimidinas/metabolismo
6.
Sci Transl Med ; 10(428)2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29444979

RESUMEN

Cancer cells adapt to their inherently increased oxidative stress through activation of the glutathione (GSH) and thioredoxin (TXN) systems. Inhibition of both of these systems effectively kills cancer cells, but such broad inhibition of antioxidant activity also kills normal cells, which is highly unwanted in a clinical setting. We therefore evaluated targeting of the TXN pathway alone and, more specifically, selective inhibition of the cytosolic selenocysteine-containing enzyme TXN reductase 1 (TXNRD1). TXNRD1 inhibitors were discovered in a large screening effort and displayed increased specificity compared to pan-TXNRD inhibitors, such as auranofin, that also inhibit the mitochondrial enzyme TXNRD2 and additional targets. For our lead compounds, TXNRD1 inhibition correlated with cancer cell cytotoxicity, and inhibitor-triggered conversion of TXNRD1 from an antioxidant to a pro-oxidant enzyme correlated with corresponding increases in cellular production of H2O2 In mice, the most specific TXNRD1 inhibitor, here described as TXNRD1 inhibitor 1 (TRi-1), impaired growth and viability of human tumor xenografts and syngeneic mouse tumors while having little mitochondrial toxicity and being better tolerated than auranofin. These results display the therapeutic anticancer potential of irreversibly targeting cytosolic TXNRD1 using small molecules and present potent and selective TXNRD1 inhibitors. Given the pronounced up-regulation of TXNRD1 in several metastatic malignancies, it seems worthwhile to further explore the potential benefit of specific irreversible TXNRD1 inhibitors for anticancer therapy.


Asunto(s)
Citosol/enzimología , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Tiorredoxina Reductasa 1/antagonistas & inhibidores , Animales , Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Peróxido de Hidrógeno/metabolismo , Masculino , Ratones SCID , Oxidación-Reducción , Relación Estructura-Actividad , Tiorredoxina Reductasa 1/química , Tiorredoxina Reductasa 1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Bioorg Med Chem Lett ; 17(23): 6489-92, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17933531

RESUMEN

Various 4-phenylpiperidine-benzoxazin-3-ones were synthesized and biologically evaluated as urotensin-II (U-II) receptor antagonists. Compound 12i was identified from in vitro evaluation as a low nanomolar antagonist against both rat and human U-II receptors. This compound showed in vivo efficacy in reversing the ear-flush response induced by U-II in rats.


Asunto(s)
Benzoxazinas/síntesis química , Piperidinas/síntesis química , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Urotensinas/metabolismo , Animales , Benzoxazinas/farmacología , Células CHO , Cricetinae , Cricetulus , Humanos , Piperidinas/farmacología , Ratas , Receptores Acoplados a Proteínas G/fisiología , Relación Estructura-Actividad , Urotensinas/antagonistas & inhibidores , Urotensinas/fisiología
8.
J Clin Endocrinol Metab ; 102(8): 2789-2797, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28609824

RESUMEN

Context: The 12-lipoxygenase (12-LO) pathway produces proinflammatory metabolites, and its activation is implicated in islet inflammation associated with type 1 and type 2 diabetes (T2D). Objectives: We aimed to test the efficacy of ML355, a highly selective, small molecule inhibitor of 12-LO, for the preservation of islet function. Design: Human islets from nondiabetic donors were incubated with a mixture of tumor necrosis factor α , interluekin-1ß, and interferon-γ to model islet inflammation. Cytokine-treated islets and human islets from T2D donors were incubated in the presence and absence of ML355. Setting: In vitro study. Participants: Human islets from organ donors aged >20 years of both sexes and any race were used. T2D status was defined from either medical history or most recent hemoglobin A1c value >6.5%. Intervention: Glucose stimulation. Main Outcome Measures: Static and dynamic insulin secretion and oxygen consumption rate (OCR). Results: ML355 prevented the reduction of insulin secretion and OCR in cytokine-treated human islets and improved both parameters in human islets from T2D donors. Conclusions: ML355 was efficacious in improving human islet function after cytokine treatment and in T2D islets in vitro. The study suggests that the blockade of the 12-LO pathway may serve as a target for both form of diabetes and provides the basis for further study of this small molecule inhibitor in vivo.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Inhibidores de la Lipooxigenasa/farmacología , Consumo de Oxígeno/efectos de los fármacos , Sulfonamidas/farmacología , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Técnicas In Vitro , Inflamación , Secreción de Insulina , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Masculino , Persona de Mediana Edad , Factor de Necrosis Tumoral alfa/farmacología , Adulto Joven
9.
J Med Chem ; 60(22): 9184-9204, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29120638

RESUMEN

We report the discovery and medicinal chemistry optimization of a novel series of pyrazole-based inhibitors of human lactate dehydrogenase (LDH). Utilization of a quantitative high-throughput screening paradigm facilitated hit identification, while structure-based design and multiparameter optimization enabled the development of compounds with potent enzymatic and cell-based inhibition of LDH enzymatic activity. Lead compounds such as 63 exhibit low nM inhibition of both LDHA and LDHB, submicromolar inhibition of lactate production, and inhibition of glycolysis in MiaPaCa2 pancreatic cancer and A673 sarcoma cells. Moreover, robust target engagement of LDHA by lead compounds was demonstrated using the cellular thermal shift assay (CETSA), and drug-target residence time was determined via SPR. Analysis of these data suggests that drug-target residence time (off-rate) may be an important attribute to consider for obtaining potent cell-based inhibition of this cancer metabolism target.


Asunto(s)
Inhibidores Enzimáticos/farmacología , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Pirazoles/farmacología , Tiazoles/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Membranas Artificiales , Ratones , Microsomas Hepáticos/efectos de los fármacos , Permeabilidad , Pirazoles/síntesis química , Pirazoles/química , Pirazoles/farmacocinética , Ratas , Solubilidad , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/química , Tiazoles/farmacocinética
11.
PLoS One ; 11(8): e0161486, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27570969

RESUMEN

The endoplasmic reticulum (ER) is involved in Ca2+ signaling and protein folding. ER Ca2+ depletion and accumulation of unfolded proteins activate the molecular chaperone GRP78 (glucose-regulated protein 78) which in turn triggers the ER stress response (ERSR) pathway aimed to restore ER homeostasis. Failure to adapt to stress, however, results in apoptosis. We and others have shown that malignant cells are more susceptible to ERSR-induced apoptosis than their normal counterparts, implicating the ERSR as a potential target for cancer therapeutics. Predicated on these findings, we developed an assay that uses a GRP78 biosensor to identify small molecule activators of ERSR in glioma cells. We performed a quantitative high-throughput screen (qHTS) against a collection of ~425,000 compounds and a comprehensive panel of orthogonal secondary assays was formulated for stringent compound validation. We identified novel activators of ERSR, including a compound with a 2,9-diazaspiro[5.5]undecane core, which depletes intracellular Ca2+ stores and induces apoptosis-mediated cell death in several cancer cell lines, including patient-derived and 3D cultures of glioma cells. This study demonstrates that our screening platform enables the identification and profiling of ERSR inducers with cytotoxic activity and advocates for characterization of these compound in in vivo models.


Asunto(s)
Alcanos/química , Alcanos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glioma/metabolismo , Animales , Apoptosis/efectos de los fármacos , Bioensayo/métodos , Western Blotting , Calcio/metabolismo , Línea Celular Tumoral , Chaperón BiP del Retículo Endoplásmico , Células HT29 , Proteínas de Choque Térmico/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos
12.
J Med Chem ; 57(19): 8099-110, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25229643

RESUMEN

Deregulation of ubiquitin conjugation or deconjugation has been implicated in the pathogenesis of many human diseases including cancer. The deubiquitinating enzyme USP1 (ubiquitin-specific protease 1), in association with UAF1 (USP1-associated factor 1), is a known regulator of DNA damage response and has been shown as a promising anticancer target. To further evaluate USP1/UAF1 as a therapeutic target, we conducted a quantitative high throughput screen of >400000 compounds and subsequent medicinal chemistry optimization of small molecules that inhibit the deubiquitinating activity of USP1/UAF1. Ultimately, these efforts led to the identification of ML323 (70) and related N-benzyl-2-phenylpyrimidin-4-amine derivatives, which possess nanomolar USP1/UAF1 inhibitory potency. Moreover, we demonstrate a strong correlation between compound IC50 values for USP1/UAF1 inhibition and activity in nonsmall cell lung cancer cells, specifically increased monoubiquitinated PCNA (Ub-PCNA) levels and decreased cell survival. Our results establish the druggability of the USP1/UAF1 deubiquitinase complex and its potential as a molecular target for anticancer therapies.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas de Arabidopsis/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Pirimidinas/síntesis química , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Pirimidinas/farmacología , Relación Estructura-Actividad , Ubiquitinación
13.
J Med Chem ; 57(2): 495-506, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24393039

RESUMEN

Human lipoxygenases (LOXs) are a family of iron-containing enzymes which catalyze the oxidation of polyunsaturated fatty acids to provide the corresponding bioactive hydroxyeicosatetraenoic acid (HETE) metabolites. These eicosanoid signaling molecules are involved in a number of physiologic responses such as platelet aggregation, inflammation, and cell proliferation. Our group has taken a particular interest in platelet-type 12-(S)-LOX (12-LOX) because of its demonstrated role in skin diseases, diabetes, platelet hemostasis, thrombosis, and cancer. Herein, we report the identification and medicinal chemistry optimization of a 4-((2-hydroxy-3-methoxybenzyl)amino)benzenesulfonamide-based scaffold. Top compounds, exemplified by 35 and 36, display nM potency against 12-LOX, excellent selectivity over related lipoxygenases and cyclooxygenases, and possess favorable ADME properties. In addition, both compounds inhibit PAR-4 induced aggregation and calcium mobilization in human platelets and reduce 12-HETE in ß-cells.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Derivados del Benceno/síntesis química , Inhibidores de la Lipooxigenasa/síntesis química , Sulfonamidas/síntesis química , Animales , Derivados del Benceno/química , Derivados del Benceno/farmacología , Disponibilidad Biológica , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Calcio/metabolismo , Humanos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/enzimología , Inhibidores de la Lipooxigenasa/química , Inhibidores de la Lipooxigenasa/farmacología , Ratones , Agregación Plaquetaria/efectos de los fármacos , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/farmacología
14.
Nat Commun ; 4: 2261, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23907321

RESUMEN

Plasmodium falciparum resistance to artemisinin derivatives, the first-line antimalarial drug, drives the search for new classes of chemotherapeutic agents. Current discovery is primarily directed against the intracellular forms of the parasite. However, late schizont-infected red blood cells (RBCs) may still rupture and cause disease by sequestration; consequently targeting invasion may reduce disease severity. Merozoite invasion of RBCs requires interaction between two parasite proteins AMA1 and RON2. Here we identify the first inhibitor of this interaction that also blocks merozoite invasion in genetically distinct parasites by screening a library of over 21,000 compounds. We demonstrate that this inhibition is mediated by the small molecule binding to AMA1 and blocking the formation of AMA1-RON complex. Electron microscopy confirms that the inhibitor prevents junction formation, a critical step in invasion that results from AMA1-RON2 binding. This study uncovers a strategy that will allow for highly effective combination therapies alongside existing antimalarial drugs.


Asunto(s)
Eritrocitos/parasitología , Malaria/parasitología , Parásitos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antimaláricos/análisis , Antimaláricos/química , Antimaláricos/farmacología , Artemisininas/farmacología , Eritrocitos/efectos de los fármacos , Eritrocitos/ultraestructura , Humanos , Concentración 50 Inhibidora , Merozoítos/efectos de los fármacos , Merozoítos/ultraestructura , Parásitos/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/química , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
15.
ACS Med Chem Lett ; 2(7): 538-43, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24900346

RESUMEN

Attenuation of fructose metabolism by the inhibition of ketohexokinase (KHK; fructokinase) should reduce body weight, free fatty acids, and triglycerides, thereby offering a novel approach to treat diabetes and obesity in response to modern diets. We have identified potent, selective inhibitors of human hepatic KHK within a series of pyrimidinopyrimidines (1). For example, 8, 38, and 47 exhibited KHK IC50 values of 12, 7, and 8 nM, respectively, and also showed potent cellular KHK inhibition (IC50 < 500 nM), which relates to their intrinsic potency vs KHK and their ability to penetrate cells. X-ray cocrystal structures of KHK complexes of 3, 8, and 47 revealed the important interactions within the enzyme's adenosine 5'-triphosphate (ATP)-binding pocket.

16.
J Med Chem ; 52(23): 7432-45, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19731961

RESUMEN

We have discovered two related chemical series of nonpeptide urotensin-II (U-II) receptor antagonists based on piperazino-phthalimide (5 and 6) and piperazino-isoindolinone (7) scaffolds. These structure types are distinctive from those of U-II receptor antagonist series reported in the literature. Antagonist 7a exhibited single-digit nanomolar potency in rat and human cell-based functional assays, as well as strong binding to the human U-II receptor. In advanced pharmacological testing, 7a blocked the effects of U-II in vitro in a rat aortic ring assay and in vivo in a rat ear-flush model. A discussion of U-II receptor antagonist pharmacophores is presented, and a specifically defined model is suggested from tricycle 13, which has a high degree of conformational constraint.


Asunto(s)
Isoindoles/química , Isoindoles/farmacología , Ftalimidas/química , Ftalimidas/farmacología , Piperazinas/química , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Células CHO , Cricetinae , Cricetulus , Ensayos Analíticos de Alto Rendimiento , Humanos , Isoindoles/síntesis química , Masculino , Ftalimidas/síntesis química , Piperazina , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA