Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(3): 563-76, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24440334

RESUMEN

The serum response factor (SRF) binds to coactivators, such as myocardin-related transcription factor-A (MRTF-A), and mediates gene transcription elicited by diverse signaling pathways. SRF/MRTF-A-dependent gene transcription is activated when nuclear MRTF-A levels increase, enabling the formation of transcriptionally active SRF/MRTF-A complexes. The level of nuclear MRTF-A is regulated by nuclear G-actin, which binds to MRTF-A and promotes its nuclear export. However, pathways that regulate nuclear actin levels are poorly understood. Here, we show that MICAL-2, an atypical actin-regulatory protein, mediates SRF/MRTF-A-dependent gene transcription elicited by nerve growth factor and serum. MICAL-2 induces redox-dependent depolymerization of nuclear actin, which decreases nuclear G-actin and increases MRTF-A in the nucleus. Furthermore, we show that MICAL-2 is a target of CCG-1423, a small molecule inhibitor of SRF/MRTF-A-dependent transcription that exhibits efficacy in various preclinical disease models. These data identify redox modification of nuclear actin as a regulatory switch that mediates SRF/MRTF-A-dependent gene transcription.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Microfilamentos/metabolismo , Oxidorreductasas/metabolismo , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Actinas/metabolismo , Secuencia de Aminoácidos , Anilidas/farmacología , Animales , Benzamidas/farmacología , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteínas de Microfilamentos/análisis , Proteínas de Microfilamentos/genética , Oxigenasas de Función Mixta/análisis , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Datos de Secuencia Molecular , Factor de Crecimiento Nervioso/metabolismo , Neuritas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Oxidación-Reducción , Oxidorreductasas/análisis , Oxidorreductasas/genética , Ratas , Alineación de Secuencia , Transactivadores , Transcripción Genética , Pez Cebra
2.
Bioorg Med Chem Lett ; 46: 128171, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34098081

RESUMEN

We have previously reported the development of indole-based CNS-active antivirals for the treatment of neurotropic alphavirus infection, but further optimization is impeded by a lack of knowledge of the molecular target and binding site. Herein we describe the design, synthesis and evaluation of a series of conformationally restricted analogues with the dual objectives of improving potency/selectivity and identifying the most bioactive conformation. Although this campaign was only modestly successful at improving potency, the sharply defined SAR of the rigid analogs enabled the definition of a three-dimensional pharmacophore, which we believe will be of value in further analog design and virtual screening for alternative antiviral leads.


Asunto(s)
Alphavirus/efectos de los fármacos , Antivirales/farmacología , Indoles/farmacología , Antivirales/síntesis química , Antivirales/química , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Indoles/síntesis química , Indoles/química , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
3.
Mol Pharmacol ; 97(6): 392-401, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32234810

RESUMEN

G protein-coupled receptor (GPCR) kinases (GRKs) play a key role in terminating signals initiated by agonist-bound GPCRs. However, chronic stimulation of GPCRs, such as that which occurs during heart failure, leads to the overexpression of GRKs and maladaptive downregulation of GPCRs on the cell surface. We previously reported the discovery of potent and selective families of GRK inhibitors based on either the paroxetine or GSK180736A scaffold. A new inhibitor, CCG258747, which is based on paroxetine, demonstrates increased potency against the GRK2 subfamily and favorable pharmacokinetic parameters in mice. CCG258747 and the closely related compound CCG258208 also showed high selectivity for the GRK2 subfamily in a kinome panel of 104 kinases. We developed a cell-based assay to screen the ability of CCG258747 and 10 other inhibitors with different GRK subfamily selectivities and with either the paroxetine or GSK180736A scaffold to block internalization of the µ-opioid receptor (MOR). CCG258747 showed the best efficacy in blocking MOR internalization among the compounds tested. Furthermore, we show that compounds based on paroxetine had much better cell permeability than those based on GSK180736A, which explains why GSK180736A-based inhibitors, although being potent in vitro, do not always show efficacy in cell-based assays. This study validates the paroxetine scaffold as the most effective for GRK inhibition in living cells, confirming that GRK2 predominantly drives internalization of MOR in the cell lines we tested and underscores the utility of high-resolution cell-based assays for assessment of compound efficacy. SIGNIFICANCE STATEMENT: G protein-coupled receptor kinases (GRKs) are attractive targets for developing therapeutics for heart failure. We have synthesized a new GRK2 subfamily-selective inhibitor, CCG258747, which has nanomolar potency against GRK2 and excellent selectivity over other kinases. A live-cell receptor internalization assay was used to test the ability of GRK2 inhibitors to impart efficacy on a GRK-dependent process in cells. Our data indicate that CCG258747 blocked the internalization of the µ-opioid receptor most efficaciously because it has the ability to cross cell membranes.


Asunto(s)
Indazoles/química , Paroxetina/química , Pirimidinas/química , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo , Animales , Western Blotting , Permeabilidad de la Membrana Celular , Cristalografía por Rayos X , Femenino , Células HEK293 , Humanos , Indazoles/farmacología , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Estructura Molecular , Pirimidinas/farmacología
4.
Nat Chem Biol ; 13(2): 218-225, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27992879

RESUMEN

The Mycobacterium tuberculosis (Mtb) DosRST two-component regulatory system promotes the survival of Mtb during non-replicating persistence (NRP). NRP bacteria help drive the long course of tuberculosis therapy; therefore, chemical inhibition of DosRST may inhibit the ability of Mtb to establish persistence and thus shorten treatment. Using a DosRST-dependent fluorescent Mtb reporter strain, a whole-cell phenotypic high-throughput screen of a ∼540,000 compound small-molecule library was conducted. The screen discovered novel inhibitors of the DosRST regulon, including three compounds that were subject to follow-up studies: artemisinin, HC102A and HC103A. Under hypoxia, all three compounds inhibit Mtb-persistence-associated physiological processes, including triacylglycerol synthesis, survival and antibiotic tolerance. Artemisinin functions by disabling the heme-based DosS and DosT sensor kinases by oxidizing ferrous heme and generating heme-artemisinin adducts. In contrast, HC103A inhibits DosS and DosT autophosphorylation activity without targeting the sensor kinase heme.


Asunto(s)
Artemisininas/farmacología , Histidina Quinasa/antagonistas & inhibidores , Mycobacterium tuberculosis/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Artemisininas/química , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Histidina Quinasa/metabolismo , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
5.
Bioorg Med Chem Lett ; 29(2): 148-154, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30528696

RESUMEN

The mitotic spindle is a microtubule-based machine that segregates a replicated set of chromosomes during cell division. Many cancer drugs alter or disrupt the microtubules that form the mitotic spindle. Microtubule-dependent molecular motors that function during mitosis are logical alternative mitotic targets for drug development. Eg5 (Kinesin-5) and Kif15 (Kinesin-12), in particular, are an attractive pair of motor proteins, as they work in concert to drive centrosome separation and promote spindle bipolarity. Furthermore, we hypothesize that the clinical failure of Eg5 inhibitors may be (in part) due to compensation by Kif15. In order to test this idea, we screened a small library of kinase inhibitors and identified GW108X, an oxindole that inhibits Kif15 in vitro. We show that GW108X has a distinct mechanism of action compared with a commercially available Kif15 inhibitor, Kif15-IN-1 and may serve as a lead with which to further develop Kif15 inhibitors as clinically relevant agents.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Cinesinas/antagonistas & inhibidores , Sondas Moleculares/farmacología , Oxindoles/farmacología , Quinazolinonas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Cinesinas/metabolismo , Sondas Moleculares/síntesis química , Sondas Moleculares/química , Estructura Molecular , Oxindoles/síntesis química , Oxindoles/química , Quinazolinonas/síntesis química , Quinazolinonas/química , Relación Estructura-Actividad
6.
J Labelled Comp Radiopharm ; 62(5): 202-208, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30828860

RESUMEN

As part of a program toward making analogues of amlexanox (1), currently under clinical investigation for the treatment of type 2 diabetes and obesity, we have synthesized derivative 5 in which deuterium has been introduced into two sites of metabolism on the C-7 isopropyl function of amlexanox. The synthesis of 5 was completed in an efficient three-step process utilizing reduction of key olefin 7b to 8 by Wilkinson's catalyst to provide specific incorporation of di-deuterium across the double bond. Compound 5 displayed nearly equivalent potency to amlexanox (IC50 , 1.1µM vs 0.6µM, respectively) against recombinant human TBK1. When incubated with human, rat, and mouse liver microsomes, amlexanox (1) and d2 -amlexanox (5) were stable (t1/2  > 60 minutes) with 1 showing marginally greater stability relative to 5 except for rat liver microsomes. These data show that incorporating deuterium into two sites of metabolism does not majorly suppress Cyp-mediated metabolism relative to amlexanox.


Asunto(s)
Aminopiridinas/síntesis química , Aminopiridinas/metabolismo , Deuterio/química , Microsomas/metabolismo , Aminopiridinas/química , Aminopiridinas/farmacología , Animales , Técnicas de Química Sintética , Estabilidad de Medicamentos , Humanos , Marcaje Isotópico , Cinética , Ratones , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas
7.
Mol Pharmacol ; 93(5): 477-488, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29476042

RESUMEN

Adhesion G protein-coupled receptors (aGPCRs) have emerged as potential therapeutic targets in multiple cancers and in neurologic diseases. However, there are few modulatory compounds that act on these receptors. The majority of aGPCRs are orphans and a general activation mechanism has only recently been defined: aGPCRs are activated by a tethered agonist. aGPCRs constitutively cleave themselves during biosynthesis to generated two-part receptors comprising an extracellular domain (ECD) and a 7-transmembrane spanning domain (7TM). ECD dissociation reveals the tethered agonist initiating G protein signaling. Synthetic peptides that mimic the tethered agonist region can activate aGPCRs. We hypothesized that small molecules could act in the same way as peptide agonists. High throughput screening of the 2000-compound Spectrum Collection library using the serum response element luciferase gene reporter assay revealed two related classes of small molecules that could activate the aGPCR GPR56/ADGRG1. The most potent compound identified was 3-α-acetoxydihydrodeoxygedunin, or 3-α-DOG. 3-α-DOG activated engineered, low-activity GPR56 7TM in independent biochemical and cell-based assays with an EC50 of ∼5 µM. The compound also activated a subset of aGPCRs but not two class A GPCRs tested. The mode of 3-α-DOG-mediated receptor activation is that of partial agonist. 3-α-DOG activated GPR56 less efficaciously than peptide agonist and could antagonize both the peptide agonist and the endogenous tethered agonist, which are pharmacological hallmarks of partial agonists. Taken together, we have uncovered a novel group of aGPCR partial agonists that will serve as invaluable resources for understanding this unique class receptors.


Asunto(s)
Receptores Acoplados a Proteínas G/agonistas , Bibliotecas de Moléculas Pequeñas , Adhesión Celular , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Limoninas/química , Unión Proteica , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Relación Estructura-Actividad
8.
Bioorg Med Chem Lett ; 28(9): 1507-1515, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29627263

RESUMEN

G protein-coupled receptor (GPCR) kinases (GRKs) regulate the desensitization and internalization of GPCRs. Two of these, GRK2 and GRK5, are upregulated in heart failure and are promising targets for heart failure treatment. Although there have been several reports of potent and selective inhibitors of GRK2 there are few for GRK5. Herein, we describe a ligand docking approach utilizing the crystal structures of the GRK2-Gßγ·GSK180736A and GRK5·CCG215022 complexes to search for amide substituents predicted to confer GRK2 and/or GRK5 potency and selectivity. From this campaign, we successfully generated two new potent GRK5 inhibitors, although neither exhibited selectivity over GRK2.


Asunto(s)
Amidas/farmacología , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Quinasa 5 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/farmacología , Amidas/síntesis química , Amidas/química , Relación Dosis-Respuesta a Droga , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 28(10): 1972-1980, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29650289

RESUMEN

The neurotropic protozoan Toxoplasma gondii is the second leading cause of death due to foodborne illness in the US, and has been designated as one of five neglected parasitic infections by the Center for Disease Control and Prevention. Currently, no treatment options exist for the chronic dormant-phase Toxoplasma infection in the central nervous system (CNS). T. gondii cathepsin L (TgCPL) has recently been implicated as a novel viable target for the treatment of chronic toxoplasmosis. In this study, we report the first body of SAR work aimed at developing potent inhibitors of TgCPL with selectivity vs the human cathepsin L. Starting from a known inhibitor of human cathepsin L, and guided by structure-based design, we were able to modulate the selectivity for Toxoplasma vs human CPL by nearly 50-fold while modifying physiochemical properties to be more favorable for metabolic stability and CNS penetrance. The overall potency of our inhibitors towards TgCPL was improved from 2 µM to as low as 110 nM and we successfully demonstrated that an optimized analog 18b is capable of crossing the BBB (0.5 brain/plasma). This work is an important first step toward development of a CNS-penetrant probe to validate TgCPL as a feasible target for the treatment of chronic toxoplasmosis.


Asunto(s)
Antiprotozoarios/química , Catepsina L/antagonistas & inhibidores , Sistema Nervioso Central/metabolismo , Dipéptidos/química , Inhibidores de Proteasas/química , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Antiprotozoarios/metabolismo , Antiprotozoarios/farmacología , Sitios de Unión , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Dominio Catalítico , Catepsina L/metabolismo , Dipéptidos/metabolismo , Dipéptidos/farmacología , Semivida , Humanos , Concentración 50 Inhibidora , Ratones , Microsomas Hepáticos/metabolismo , Simulación de Dinámica Molecular , Permeabilidad/efectos de los fármacos , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Proteínas Protozoarias/metabolismo , Relación Estructura-Actividad , Toxoplasma/efectos de los fármacos , Toxoplasma/enzimología
10.
J Nanobiotechnology ; 16(1): 97, 2018 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-30482196

RESUMEN

BACKGROUND: Sustained drug delivery is a large unmet clinical need in glaucoma. Here, we incorporated a Myocardin-Related Transcription Factor/Serum Response Factor inhibitor, CCG-222740, into slow release large unilamellar vesicles derived from the liposomes DOTMA (1,2-di-O-octadecenyl-3-trimethylammonium propane) and DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine), and tested their effects in vitro and in vivo. RESULTS: The vesicles were spherical particles of around 130 nm and were strongly cationic. A large amount of inhibitor could be incorporated into the vesicles. We showed that the nanocarrier CCG-222740 formulation gradually released the inhibitor over 14 days using high performance liquid chromatography. Nanocarrier CCG-222740 significantly decreased ACTA2 gene expression and was not cytotoxic in human conjunctival fibroblasts. In vivo, nanocarrier CCG-222740 doubled the bleb survival from 11.0 ± 0.6 days to 22.0 ± 1.3 days (p = 0.001), decreased conjunctival scarring and did not have any local or systemic adverse effects in a rabbit model of glaucoma filtration surgery. CONCLUSIONS: Our study demonstrates proof-of-concept that a nanocarrier-based formulation efficiently achieves a sustained release of a Myocardin-Related Transcription Factor/Serum Response Factor inhibitor and prevents conjunctival fibrosis in an established rabbit model of glaucoma filtration surgery.


Asunto(s)
Preparaciones de Acción Retardada/química , Sistemas de Liberación de Medicamentos , Factor de Respuesta Sérica/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Enfermedades de la Conjuntiva/tratamiento farmacológico , Femenino , Fibroblastos/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Humanos , Liposomas/química , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Conejos , Distribución Tisular , Transactivadores/antagonistas & inhibidores , Transactivadores/química
11.
Mol Pharmacol ; 92(6): 707-717, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29070696

RESUMEN

G protein-coupled receptor kinases (GRKs) phosphorylate activated receptors to promote arrestin binding, decoupling from heterotrimeric G proteins, and internalization. GRK2 and GRK5 are overexpressed in the failing heart and thus have become therapeutic targets. Previously, we discovered two classes of GRK2-selective inhibitors, one stemming from GSK180736A, a Rho-associated coiled-coil containing kinase 1 (ROCK1) inhibitor, the other from paroxetine, a selective serotonin-reuptake inhibitor. These two classes of compounds bind to the GRK2 active site in a similar configuration but contain different hinge-binding "warheads": indazole and benzodioxole, respectively. We surmised from our prior studies that an indazole would be the stronger hinge binder and would impart increased potency when substituted for benzodioxole in paroxetine derivatives. To test this hypothesis, we synthesized a series of hybrid compounds that allowed us to compare the effects of inhibitors that differ only in the identity of the warhead. The indazole-paroxetine analogs were indeed more potent than their respective benzodioxole derivatives but lost selectivity. To investigate how these two warheads dictate selectivity, we determined the crystal structures of three of the indazole hybrid compounds (CCG224061, CCG257284, and CCG258748) in complex with GRK2-Gßγ Comparison of these structures with those of analogous benzodioxole-containing complexes confirmed that the indazole-paroxetine hybrids form stronger interactions with the hinge of the kinase but also stabilize a distinct conformation of the kinase domain of GRK2 compared with previous complexes with paroxetine analogs. This conformation is analogous to one that can be assumed by GRK5, at least partially explaining the loss in selectivity.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Quinasa 5 del Receptor Acoplado a Proteína-G/farmacología , Indazoles/farmacología , Paroxetina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Quinasa 2 del Receptor Acoplado a Proteína-G/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Inhibidores Selectivos de la Recaptación de Serotonina , Quinasas Asociadas a rho/metabolismo
12.
Bioorg Med Chem Lett ; 27(8): 1744-1749, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28285914

RESUMEN

We recently reported the development of a novel inhibitor of Rho-mediated gene transcription (1, CCG-203971) that is efficacious in multiple animal models of acute fibrosis, including scleroderma, when given intraperitoneally. The modest in vivo potency and poor pharmacokinetics (PK) of this lead, however, make it unsuitable for long term efficacy studies. We therefore undertook a systematic medicinal chemistry effort to improve both the metabolic stability and the solubility of 1, resulting in the identification of two analogs achieving over 10-fold increases in plasma exposures in mice. We subsequently showed that one of these analogs (8f, CCG-232601) could inhibit the development of bleomycin-induced dermal fibrosis in mice when administered orally at 50mg/kg, an effect that was comparable to what we had observed earlier with 1 at a 4-fold higher IP dose.


Asunto(s)
Ácidos Nipecóticos/farmacocinética , Ácidos Nipecóticos/uso terapéutico , Factor Rho/antagonistas & inhibidores , Esclerodermia Sistémica/tratamiento farmacológico , Piel/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Administración Oral , Animales , Modelos Animales de Enfermedad , Fibrosis , Células HEK293 , Humanos , Ratones , Ácidos Nipecóticos/administración & dosificación , Ácidos Nipecóticos/química , Factor Rho/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Elemento de Respuesta al Suero/efectos de los fármacos , Piel/metabolismo , Piel/patología , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo
13.
J Biol Chem ; 290(34): 20649-20659, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26032411

RESUMEN

G protein-coupled receptor kinases (GRKs) regulate cell signaling by initiating the desensitization of active G protein-coupled receptors. The two most widely expressed GRKs (GRK2 and GRK5) play a role in cardiovascular disease and thus represent important targets for the development of novel therapeutic drugs. In the course of a GRK2 structure-based drug design campaign, one inhibitor (CCG215022) exhibited nanomolar IC50 values against both GRK2 and GRK5 and good selectivity against other closely related kinases such as GRK1 and PKA. Treatment of murine cardiomyocytes with CCG215022 resulted in significantly increased contractility at 20-fold lower concentrations than paroxetine, an inhibitor with more modest selectivity for GRK2. A 2.4 Å crystal structure of the GRK5·CCG215022 complex was determined and revealed that the inhibitor binds in the active site similarly to its parent compound GSK180736A. As designed, its 2-pyridylmethyl amide side chain occupies the hydrophobic subsite of the active site where it forms three additional hydrogen bonds, including one with the catalytic lysine. The overall conformation of the GRK5 kinase domain is similar to that of a previously determined structure of GRK6 in what is proposed to be its active state, but the C-terminal region of the enzyme adopts a distinct conformation. The kinetic properties of site-directed mutants in this region are consistent with the hypothesis that this novel C-terminal structure is representative of the membrane-bound conformation of the enzyme.


Asunto(s)
Fármacos Cardiovasculares/química , Inhibidores Enzimáticos/química , Quinasa 5 del Receptor Acoplado a Proteína-G/química , Miocitos Cardíacos/efectos de los fármacos , Piridinas/química , Animales , Fármacos Cardiovasculares/síntesis química , Fármacos Cardiovasculares/farmacología , Dominio Catalítico , Bovinos , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/aislamiento & purificación , Expresión Génica , Tabiques Cardíacos/química , Tabiques Cardíacos/citología , Tabiques Cardíacos/efectos de los fármacos , Tabiques Cardíacos/enzimología , Ventrículos Cardíacos/química , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/enzimología , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/química , Miocitos Cardíacos/citología , Miocitos Cardíacos/enzimología , Paroxetina/química , Paroxetina/farmacología , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Piridinas/síntesis química , Piridinas/farmacología , Alineación de Secuencia
14.
Am J Pathol ; 185(4): 969-86, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25681733

RESUMEN

Myofibroblasts are crucial to the pathogenesis of tissue fibrosis. Their formation of stress fibers results in the release of myocardin-related transcription factor (MRTF), a transcriptional coactivator of serum response factor (SRF). MRTF-A (Mkl1)-deficient mice are protected from lung fibrosis. We hypothesized that the SRF/MRTF pathway inhibitor CCG-203971 would modulate myofibroblast function in vitro and limit lung fibrosis in vivo. Normal and idiopathic pulmonary fibrosis lung fibroblasts were treated with/without CCG-203971 (N-[4-chlorophenyl]-1-[3-(2-furanyl)benzoyl]-3-piperidine carboxamide) and/or Fas-activating antibody in the presence/absence of transforming growth factor (TGF)-ß1, and apoptosis was assessed. In vivo studies examined the effect of therapeutically administered CCG-203971 on lung fibrosis in two distinct murine models of fibrosis induced by bleomycin or targeted type II alveolar epithelial injury. In vitro, CCG-203971 prevented nuclear localization of MRTF-A; increased the apoptotic susceptibility of normal and idiopathic pulmonary fibrosis fibroblasts; blocked TGF-ß1-induced myofibroblast differentiation; and inhibited TGF-ß1-induced expression of fibronectin, X-linked inhibitor of apoptosis, and plasminogen activator inhibitor-1. TGF-ß1 did not protect fibroblasts or myofibroblasts from apoptosis in the presence of CCG-203971. In vivo, CCG-203971 significantly reduced lung collagen content in both murine models while decreasing alveolar plasminogen activator inhibitor-1 and promoting myofibroblast apoptosis. These data support a central role of the SRF/MRTF pathway in the pathobiology of lung fibrosis and suggest that its inhibition can help resolve lung fibrosis by promoting fibroblast apoptosis.


Asunto(s)
Apoptosis , Pulmón/metabolismo , Pulmón/patología , Mesodermo/patología , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Adulto , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoprotección/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis , Humanos , Inflamación/patología , Mesodermo/efectos de los fármacos , Ratones Endogámicos C57BL , Miofibroblastos/patología , Ácidos Nipecóticos/administración & dosificación , Ácidos Nipecóticos/farmacología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sus scrofa , Factor de Crecimiento Transformador beta1/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Receptor fas/metabolismo
15.
Pediatr Surg Int ; 32(12): 1103-1114, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27670279

RESUMEN

BACKGROUND: Blockade of the renin-angiotensin system (RAS) has been shown to alleviate inflammatory processes in the gastrointestinal tract. The aim of this study was to determine if blockade of the RAS would be effective in an immunologically relevant colitis model, and to compare outcome with an acute colitis model. METHODS: A losartan analog, CCG-203025 (C23H26ClN3O5S) containing a highly polar sulfonic acid moiety that we expected would allow localized mucosal antagonism with minimal systemic absorption was selected as an angiotensin II type 1a receptor antagonist (AT1aR-A). Two colitis models were studied: (1) Acute colitis was induced in 8- to 10-week-old C57BL/6J mice by 2.5 % dextran sodium sulfate (DSS, in drinking water) for 7 days. (2) IL10-/-colitis Piroxicam (200 ppm) was administered orally in feed to 5-week-old IL-10-/-mice (C57BL/6J background) for 14 days followed by enalaprilat (ACE-I), CCG-203025 or PBS administered transanally for 14 days. RESULTS: In the DSS model, weight loss and histologic score for CCG-203025 were better than with placebo. In the IL10-/-model, ACE-I suppressed histologic damage better than CCG-203025. Both ACE-I and CCG-203025 reduced pro-inflammatory cytokines and chemokines. CONCLUSIONS: This study demonstrated the therapeutic efficacy of both ACE-I and AT1aR-A for preventing the development of both acute and immunologically relevant colitis.


Asunto(s)
Colitis/inmunología , Colitis/prevención & control , Losartán/análogos & derivados , Losartán/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Enfermedad Aguda , Bloqueadores del Receptor Tipo 1 de Angiotensina II/inmunología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/inmunología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Colitis/patología , Inhibidores de la Ciclooxigenasa/inmunología , Inhibidores de la Ciclooxigenasa/farmacología , Modelos Animales de Enfermedad , Enalaprilato/inmunología , Enalaprilato/farmacología , Losartán/inmunología , Ratones , Ratones Endogámicos C57BL , Piroxicam/inmunología , Piroxicam/farmacología , Sistema Renina-Angiotensina/inmunología
16.
J Virol ; 88(20): 11825-33, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100850

RESUMEN

To combat emerging coronaviruses, developing safe and efficient platforms to evaluate viral protease activities and the efficacy of protease inhibitors is a high priority. Here, we exploit a biosafety level 2 (BSL-2) chimeric Sindbis virus system to evaluate protease activities and the efficacy of inhibitors directed against the papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus (SARS-CoV), a biosafety level 3 (BSL-3) pathogen. We engineered Sindbis virus to coexpress PLpro and a substrate, murine interferon-stimulated gene 15 (ISG15), and found that PLpro mediates removal of ISG15 (deISGylation) from cellular proteins. Mutation of the catalytic cysteine residue of PLpro or addition of a PLpro inhibitor blocked deISGylation in virus-infected cells. Thus, deISGylation is a marker of PLpro activity. Infection of alpha/beta interferon receptor knockout (IFNAR(-/-)) mice with these chimeric viruses revealed that PLpro deISGylation activity removed ISG15-mediated protection during viral infection. Importantly, administration of a PLpro inhibitor protected these mice from lethal infection, demonstrating the efficacy of a coronavirus protease inhibitor in a mouse model. However, this PLpro inhibitor was not sufficient to protect the mice from lethal infection with SARS-CoV MA15, suggesting that further optimization of the delivery and stability of PLpro inhibitors is needed. We extended the chimeric-virus platform to evaluate the papain-like protease/deISGylating activity of Middle East respiratory syndrome coronavirus (MERS-CoV) to provide a small-animal model to evaluate PLpro inhibitors of this recently emerged pathogen. This platform has the potential to be universally adaptable to other viral and cellular enzymes that have deISGylating activities. Importance: Evaluating viral protease inhibitors in a small-animal model is a critical step in the path toward antiviral drug development. We modified a biosafety level 2 chimeric virus system to facilitate evaluation of inhibitors directed against highly pathogenic coronaviruses. We used this system to demonstrate the in vivo efficacy of an inhibitor of the papain-like protease of severe acute respiratory syndrome coronavirus. Furthermore, we demonstrate that the chimeric-virus system can be adapted to study the proteases of emerging human pathogens, such as Middle East respiratory syndrome coronavirus. This system provides an important tool to rapidly assess the efficacy of protease inhibitors targeting existing and emerging human pathogens, as well as other enzymes capable of removing ISG15 from cellular proteins.


Asunto(s)
Coronavirus/fisiología , Modelos Animales de Enfermedad , Papaína/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Chlorocebus aethiops , Coronavirus/enzimología , Cricetinae , Ratones , Células Vero
17.
J Virol ; 88(19): 11199-214, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25031353

RESUMEN

Neurotropic alphaviruses, including western, eastern, and Venezuelan equine encephalitis viruses, cause serious and potentially fatal central nervous system infections in humans for which no currently approved therapies exist. We previously identified a series of thieno[3,2-b]pyrrole derivatives as novel inhibitors of neurotropic alphavirus replication, using a cell-based phenotypic assay (W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275), and subsequently developed second- and third-generation indole-2-carboxamide derivatives with improved potency, solubility, and metabolic stability (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). In this report, we describe the antiviral activity of the most promising third-generation lead compound, CCG205432, and closely related analogs CCG206381 and CCG209023. These compounds have half-maximal inhibitory concentrations of ∼1 µM and selectivity indices of >100 in cell-based assays using western equine encephalitis virus replicons. Furthermore, CCG205432 retains similar potency against fully infectious virus in cultured human neuronal cells. These compounds show broad inhibitory activity against a range of RNA viruses in culture, including members of the Togaviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Although their exact molecular target remains unknown, mechanism-of-action studies reveal that these novel indole-based compounds target a host factor that modulates cap-dependent translation. Finally, we demonstrate that both CCG205432 and CCG209023 dampen clinical disease severity and enhance survival of mice given a lethal western equine encephalitis virus challenge. These studies demonstrate that indole-2-carboxamide compounds are viable candidates for continued preclinical development as inhibitors of neurotropic alphaviruses and, potentially, of other RNA viruses. IMPORTANCE There are currently no approved drugs to treat infections with alphaviruses. We previously identified a novel series of compounds with activity against these potentially devastating pathogens (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). We have now produced third-generation compounds with enhanced potency, and this manuscript provides detailed information on the antiviral activity of these advanced-generation compounds, including activity in an animal model. The results of this study represent a notable achievement in the continued development of this novel class of antiviral inhibitors.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Equina del Oeste/efectos de los fármacos , Encefalomielitis Equina/tratamiento farmacológico , Indoles/farmacología , Piridinas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/síntesis química , Bunyaviridae/efectos de los fármacos , Bunyaviridae/crecimiento & desarrollo , Línea Celular , Virus de la Encefalitis Equina del Oeste/crecimiento & desarrollo , Virus de la Encefalitis Equina del Oeste/patogenicidad , Encefalomielitis Equina/mortalidad , Encefalomielitis Equina/virología , Femenino , Indoles/síntesis química , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/virología , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/crecimiento & desarrollo , Picornaviridae/efectos de los fármacos , Picornaviridae/crecimiento & desarrollo , Biosíntesis de Proteínas/efectos de los fármacos , Piridinas/síntesis química , Replicón/efectos de los fármacos , Relación Estructura-Actividad , Análisis de Supervivencia
18.
Bioorg Med Chem ; 23(7): 1569-87, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25740634

RESUMEN

Neurotropic alphaviruses are debilitating pathogens that infect the central nervous system (CNS) and are transmitted to humans via mosquitoes. There exist no effective human vaccines against these viruses, underlining the need for effective antivirals, but no antiviral drugs are available for treating infection once the viruses have invaded the CNS. Previously, we reported the development of novel indole-2-carboxamide-based inhibitors of alphavirus replication that demonstrate significant reduction of viral titer and achieve measurable brain permeation in a pharmacokinetic mouse model. Herein we report our continued efforts to improve physicochemical properties predictive of in vivo blood-brain barrier (BBB) permeability through reduction of overall molecular weight, replacing the indole core with a variety of aromatic and non-aromatic monocyclics. These studies culminated in the identification of simple anthranilamides that retain excellent potency with improved metabolic stability and significantly greater aqueous solubility. Furthermore, in a live virus study, we showed that two new compounds were capable of reducing viral titer by two orders of magnitude and that these compounds likely exert their effects through a mechanism similar to that of our indole-2-carboxamide inhibitors.


Asunto(s)
Alphavirus/efectos de los fármacos , Antivirales/farmacología , Descubrimiento de Drogas/métodos , Replicación Viral/efectos de los fármacos , ortoaminobenzoatos/farmacología , Alphavirus/fisiología , Animales , Antivirales/química , Ratones , Ratones Endogámicos BALB C , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/virología , Replicación Viral/fisiología , ortoaminobenzoatos/química
19.
Proc Natl Acad Sci U S A ; 109(9): 3469-74, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22331877

RESUMEN

The widespread occurrence of antibiotic resistance among human pathogens is a major public health problem. Conventional antibiotics typically target bacterial killing or growth inhibition, resulting in strong selection for the development of antibiotic resistance. Alternative therapeutic approaches targeting microbial pathogenicity without inhibiting growth might minimize selection for resistant organisms. Compounds inhibiting gene expression of streptokinase (SK), a critical group A streptococcal (GAS) virulence factor, were identified through a high-throughput, growth-based screen on a library of 55,000 small molecules. The lead compound [Center for Chemical Genomics 2979 (CCG-2979)] and an analog (CCG-102487) were confirmed to also inhibit the production of active SK protein. Microarray analysis of GAS grown in the presence of CCG-102487 showed down-regulation of a number of important virulence factors in addition to SK, suggesting disruption of a general virulence gene regulatory network. CCG-2979 and CCG-102487 both enhanced granulocyte phagocytosis and killing of GAS in an in vitro assay, and CCG-2979 also protected mice from GAS-induced mortality in vivo. These data suggest that the class of compounds represented by CCG-2979 may be of therapeutic value for the treatment of GAS and potentially other gram-positive infections in humans.


Asunto(s)
Antibacterianos/uso terapéutico , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Quinazolinas/uso terapéutico , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes/efectos de los fármacos , Estreptoquinasa/antagonistas & inhibidores , Animales , Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , Depresión Química , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Especificidad del Huésped/genética , Humanos , Resistencia a la Kanamicina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estructura Molecular , Fagocitosis/efectos de los fármacos , Plasminógeno/genética , Regiones Promotoras Genéticas/genética , Quinazolinas/aislamiento & purificación , Quinazolinas/farmacología , Bibliotecas de Moléculas Pequeñas , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Estreptoquinasa/biosíntesis , Estreptoquinasa/genética , Virulencia/efectos de los fármacos , Virulencia/genética
20.
J Lipid Res ; 55(7): 1215-25, 2014 07.
Artículo en Inglés | MEDLINE | ID: mdl-24534703

RESUMEN

Glycosphingolipid (GSL) storage diseases have been the focus of efforts to develop small molecule therapeutics from design, experimental proof of concept studies, and clinical trials. Two primary alternative strategies that have been pursued include pharmacological chaperones and GSL synthase inhibitors. There are theoretical advantages and disadvantages to each of these approaches. Pharmacological chaperones are specific for an individual glycoside hydrolase and for the specific mutation present, but no candidate chaperone has been demonstrated to be effective for all mutations leading to a given disorder. Synthase inhibitors target single enzymes such as glucosylceramide synthase and inhibit the formation of multiple GSLs. A glycolipid synthase inhibitor could potentially be used to treat multiple diseases, but at the risk of lowering nontargeted cellular GSLs that are important for normal health. The basis for these strategies and specific examples of compounds that have led to clinical trials is the focus of this review.


Asunto(s)
Inhibidores de Glicósido Hidrolasas/uso terapéutico , Glicósido Hidrolasas , Glicoesfingolípidos , Enfermedades por Almacenamiento Lisosomal , Chaperonas Moleculares/uso terapéutico , Mutación , Animales , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Glicoesfingolípidos/genética , Glicoesfingolípidos/metabolismo , Humanos , Enfermedades por Almacenamiento Lisosomal/tratamiento farmacológico , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/metabolismo , Enfermedades por Almacenamiento Lisosomal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA