Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Bioorg Med Chem ; 28(11): 115480, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32327351

RESUMEN

Studies demonstrate that small molecule targeting of atypical protein kinase C (aPKC) may provide an effective means to control vascular permeability, prevent edema, and reduce inflammation providing novel and important alternatives to anti-VEGF therapies for certain blinding eye diseases. Based on a literature tricyclic thieno[2,3-d]pyrimidine lead (1), an ATP-competitive inhibitor of the aPKC iota (ι) and aPKC zeta (ζ) isoforms, we have synthesized a small series of compounds in 1-2 steps from a readily available chloro intermediate. A single pyridine congener was also made using 2D NMR to assign regiochemistry. Within the parent pyrimidine series, a range of potencies was observed against aPKCζ whereas the pyridine congener was inactive. Selected compounds were also tested for their effect toward VEGF-induced permeability in BREC cells. The most potent of these (7l) was further assayed against the aPKCι isoform and showed a favorable selectivity profile against a panel of 31 kinases, including kinases from the AGC superfamily, with a focus on PKC isoforms and kinases previously shown to affect permeability. Further testing of 7l in a luciferase assay in HEK293 cells showed an ability to prevent TNF-α induced NFκB activation while not having any effect on cell survival. Intravitreal administration of 7l to the eye yielded a complete reduction in permeability in a test to determine whether the compound could block VEGF- and TNFα-induced permeability across the retinal vasculature in a rat model. The compound in mice displayed good microsomal stability and in plasma moderate exposure (AUC and Cmax), low clearance, a long half-life and high oral bioavailability. With IV dosing, higher levels were observed in the brain and eye relative to plasma, with highest levels in the eye by either IV or PO dosing. With a slow oral absorption profile, 7l accumulates in the eye to maintain a high concentration after dosing with higher levels than in plasma. Compound 7l may represent a class of aPKC inhibitors for further investigation.


Asunto(s)
Citocinas/antagonistas & inhibidores , Edema/tratamiento farmacológico , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Edema/inducido químicamente , Edema/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Estructura Molecular , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/síntesis química , Pirimidinas/química , Ratas , Ratas Long-Evans , Relación Estructura-Actividad , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Molecules ; 25(18)2020 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-32927749

RESUMEN

Nitroimidazole drugs have a long history as therapeutic agents to treat bacterial and parasitic diseases. The discovery in 1989 of a bicyclic nitroimidazole lead, displaying in vitro and in vivo antitubercular activity, spurred intensive exploration of this and related scaffolds, which led to the regulatory approval of pretomanid and delamanid as a new class of tuberculosis drugs. Much of the discovery work related to this took place over a 20-year period ending in 2010, which is covered in a number of cited reviews. This review highlights subsequent research published over the 2011-August 2020 timeframe, and captures detailed structure-activity relationship studies and synthetic strategies directed towards uncovering newer generation drugs for both tuberculosis and selected neglected tropical diseases. Additionally, this review presents in silico calculations relating to the drug-like properties of lead compounds and clinical agents, as well as chemical development and manufacturing processes toward providing bulk drug supplies.


Asunto(s)
Antituberculosos/química , Antituberculosos/farmacología , Desarrollo de Medicamentos , Mycobacterium tuberculosis/efectos de los fármacos , Nitroimidazoles/química , Nitroimidazoles/farmacología , Antituberculosos/uso terapéutico , Técnicas de Química Sintética , Diseño de Fármacos , Humanos , Nitroimidazoles/uso terapéutico , Relación Estructura-Actividad , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología
3.
Tetrahedron Lett ; 60(30): 2035-2037, 2019 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32831416

RESUMEN

We report on an approach to truncate the tricyclic 5H-chromeno[2,3-b]pyridin-5-one core of amlexanox, an approved drug under investigation for the treatment of obesity, to the bicyclic 4H-pyrano[2,3-b]pyridin-4-one (8-azachromone) core. A short, concise synthesis generates a key intermediate with requisite functionality on the pyridyl A-ring and iodo functionality on the 4-pyrone B-ring upon which palladium-catalyzed cross-coupling and subsequent reactions generate representative analogues. One of these shows a 14.2-fold increase in aqueous solubility over amlexanox.

4.
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
5.
Mol Pharmacol ; 94(4): 1210-1219, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30082428

RESUMEN

Chronic low-grade inflammation is a hallmark of obesity, which is a risk factor for the development of type 2 diabetes. The drug amlexanox inhibits IκB kinase ε (IKKε) and TANK binding kinase 1 (TBK1) to promote energy expenditure and improve insulin sensitivity. Clinical studies have demonstrated efficacy in a subset of diabetic patients with underlying adipose tissue inflammation, albeit with moderate potency, necessitating the need for improved analogs. Herein we report crystal structures of TBK1 in complex with amlexanox and a series of analogs that modify its carboxylic acid moiety. Removal of the carboxylic acid or mutation of the adjacent Thr156 residue significantly reduces potency toward TBK1, whereas conversion to a short amide or ester nearly abolishes the inhibitory effects. IKKε is less affected by these modifications, possibly due to variation in its hinge that allows for increased conformational plasticity. Installation of a tetrazole carboxylic acid bioisostere improved potency to 200 and 400 nM toward IKKε and TBK1, respectively. Despite improvements in the in vitro potency, no analog produced a greater response in adipocytes than amlexanox, perhaps because of altered absorption and distribution. The structure-activity relationships and cocrystal structures described herein will aid in future structure-guided inhibitor development using the amlexanox pharmacophore for the treatment of obesity and type 2 diabetes.


Asunto(s)
Aminopiridinas/farmacología , Ácidos Carboxílicos/farmacología , Quinasa I-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad
6.
Org Biomol Chem ; 16(37): 8245-8248, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-29537042

RESUMEN

Two novel cyclic quaternary amine crosslinking probes are synthesized for structural mass spectrometry of protein complexes in solution and for analysis of protein interactions in organellar and whole cell extracts. Each exhibits high aqueous solubility, excellent protein crosslinking efficiencies, low collision induced dissociation (CID) energy fragmentation efficiencies, high stoichiometries of reaction, increased charges of crosslinked peptide ions, and maintenance of overall surface charge balance of crosslinked proteins.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Proteínas/química , Compuestos de Amonio Cuaternario/química , Espectrometría de Masas en Tándem/métodos , Secuencia de Aminoácidos , Animales , Fructosa-Bifosfato Aldolasa/química , Humanos , Iones/química , Modelos Moleculares , Péptidos/análisis
7.
Bioorg Med Chem ; 26(20): 5443-5461, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30270002

RESUMEN

The non-canonical IκB kinases TANK-binding kinase 1 (TBK1) and inhibitor of nuclear factor kappa-B kinase ε (IKKε) play a key role in insulin-independent pathways that promote energy storage and block adaptive energy expenditure during obesity. Utilizing docking calculations and the x-ray structure of TBK1 bound to amlexanox, an inhibitor of these kinases with modest potency, a series of analogues was synthesized to develop a structure activity relationship (SAR) around the A- and C-rings of the core scaffold. A strategy was developed wherein R7 and R8 A-ring substituents were incorporated late in the synthetic sequence by utilizing palladium-catalyzed cross-coupling reactions on appropriate bromo precursors. Analogues display IC50 values as low as 210 nM and reveal A-ring substituents that enhance selectivity toward either kinase. In cell assays, selected analogues display enhanced phosphorylation of p38 or TBK1 and elicited IL-6 secretion in 3T3-L1 adipocytes better than amlexanox. An analogue bearing a R7 cyclohexyl modification demonstrated robust IL-6 production in 3T3-L1 cells as well as a phosphorylation marker of efficacy and was tested in obese mice where it promoted serum IL-6 response, weight loss, and insulin sensitizing effects comparable to amlexanox. These studies provide impetus to expand the SAR around the amlexanox core toward uncovering analogues with development potential.


Asunto(s)
Quinasa I-kappa B/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/química , Piridinas/farmacología , Células 3T3-L1 , Aminación , Animales , Fármacos Antiobesidad/síntesis química , Fármacos Antiobesidad/química , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/uso terapéutico , Cromanos/síntesis química , Cromanos/química , Cromanos/farmacología , Cromanos/uso terapéutico , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Quinasa I-kappa B/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Obesidad/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/metabolismo , Piridinas/síntesis química , Piridinas/uso terapéutico
8.
Blood ; 125(23): 3588-97, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25814533

RESUMEN

Usp9x was recently shown to be highly expressed in myeloma patients with short progression-free survival and is proposed to enhance stability of the survival protein Mcl-1. In this study, we found that the partially selective Usp9x deubiquitinase inhibitor WP1130 induced apoptosis and reduced Mcl-1 protein levels. However, short hairpin RNA-mediated knockdown (KD) of Usp9x in myeloma cells resulted in transient induction of apoptosis, followed by a sustained reduction in cell growth. A compensatory upregulation of Usp24, a deubiquitinase closely related to Usp9x, in Usp9x KD cells was noted. Direct Usp24 KD resulted in marked induction of myeloma cell death that was associated with a reduction of Mcl-1. Usp24 was found to sustain myeloma cell survival and Mcl-1 regulation in the absence of Usp9x. Both Usp9x and Usp24 were expressed and activated in primary myeloma cells whereas Usp24 protein overexpression was noted in some patients with drug-refractory myeloma and other B-cell malignancies. Furthermore, we improved the drug-like properties of WP1130 and demonstrated that the novel compound EOAI3402143 dose-dependently inhibited Usp9x and Usp24 activity, increased tumor cell apoptosis, and fully blocked or regressed myeloma tumors in mice. We conclude that small-molecule Usp9x/Usp24 inhibitors may have therapeutic activity in myeloma.


Asunto(s)
Apoptosis/efectos de los fármacos , Cianoacrilatos/farmacología , Inhibidores Enzimáticos/farmacología , Linfoma de Células del Manto/tratamiento farmacológico , Mieloma Múltiple/tratamiento farmacológico , Piridinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Animales , Apoptosis/genética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfoma de Células del Manto/enzimología , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/patología , Masculino , Ratones , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
9.
Chemistry ; 23(56): 13875-13878, 2017 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-28833638

RESUMEN

Brequinar, a potent dihydroorotate dehydrogenase (DHODH) inhibitor, has been evaluated in multiple clinical trials as a potential treatment for cancer. To further understand brequinar-based DHODH inhibition and DHODH's therapeutic relevance in cancer, we have developed novel brequinar-based probes. We disclose a 16-step convergent synthesis of the first brequinar-PROTAC and a four-step approach towards the first mitochondrial-directed brequinar probe. A PROTAC and mitochondria-directed probe of brequinar both possess cytotoxicity that is superior to brequinar in a colony formation assay.

10.
Neurochem Res ; 42(6): 1823-1832, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28255754

RESUMEN

Glutamate uptake into synaptic vesicles in nerve terminals is a pivotal step in glutamate synaptic transmission. Glutamate is the major excitatory neurotransmitter and, as such, the vesicular glutamate transporter (VGLUT) responsible for this uptake is involved in a variety of nervous system functions and various types of pathophysiology. As yet, no VGLUT-specific, membrane-permeable agents have been developed to affect neuronal function in intact neurons, although two potent VGLUTspecific inhibitors are known. These compounds contain diazo and highly charged sulfonic acid groups, rendering them membrane-impermeable and potentially cytotoxic. In an effort to eliminate these undesirable properties, we have developed two novel agents, Brilliant Yellow analogs 1 and 2, which are free of these two groups. We show here that these agents retain highly VGLUT-selective inhibitory activity, despite their reduction in potency, and exhibit no significant cellular toxicity. Potential use of this molecular modification is discussed.


Asunto(s)
Compuestos Azo/química , Compuestos Azo/metabolismo , Bencenosulfonatos/química , Bencenosulfonatos/metabolismo , Proteínas de Transporte Vesicular de Glutamato/análisis , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Encéfalo/metabolismo , Química Encefálica/fisiología , Células PC12 , Ratas , Vesículas Sinápticas/química , Vesículas Sinápticas/metabolismo
11.
Bioorg Med Chem Lett ; 27(18): 4350-4353, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28838693

RESUMEN

Starting from a known non-specific agonist (1) of nicotinic acetylcholine receptors (nAChRs), rationally guided structural-based design resulted in the discovery of a small series of 5'-phenyl-1,2,5,6-tetrahydro-3,3'-bipyridines (3a-3e) incorporating a phenyl ring off the pyridine core of 1. The compounds were synthesized via successive Suzuki couplings on a suitably functionalized pyridine starting monomer 4 to append phenyl and pyridyl substituents off the 3- and 5-positions, respectively, and then subsequent modifications were made on the flanking pyridyl ring to provide target compounds. Compound 3a is a novel antagonist, which is highly selective for α3ß4 nAChR (Ki=123nM) over the α4ß2 and α7 receptors.


Asunto(s)
Diseño de Fármacos , Antagonistas Nicotínicos/farmacología , Piridinas/farmacología , Receptores Nicotínicos/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Antagonistas Nicotínicos/síntesis química , Antagonistas Nicotínicos/química , Piridinas/síntesis química , Piridinas/química , Ratas , Relación Estructura-Actividad
12.
Antimicrob Agents Chemother ; 60(7): 4183-96, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27139470

RESUMEN

Due to the rise of antibiotic resistance and the small number of effective antiviral drugs, new approaches for treating infectious diseases are urgently needed. Identifying targets for host-based therapies represents an emerging strategy for drug discovery. The ubiquitin-proteasome system is a central mode of signaling in the eukaryotic cell and may be a promising target for therapies that bolster the host's ability to control infection. Deubiquitinase (DUB) enzymes are key regulators of the host inflammatory response, and we previously demonstrated that a selective DUB inhibitor and its derivative promote anti-infective activities in host cells. To find compounds with anti-infective efficacy but improved toxicity profiles, we tested a library of predominantly 2-cyano-3-acrylamide small-molecule DUB inhibitors for anti-infective activity in macrophages against two intracellular pathogens: murine norovirus (MNV) and Listeria monocytogenes We identified compound C6, which inhibited DUB activity in human and murine cells and reduced intracellular replication of both pathogens with minimal toxicity in cell culture. Treatment with C6 did not significantly affect the ability of macrophages to internalize virus, suggesting that the anti-infective activity interferes with postentry stages of the MNV life cycle. Metabolic stability and pharmacokinetic assays showed that C6 has a half-life in mouse liver microsomes of ∼20 min and has a half-life of approximately 4 h in mice when administered intravenously. Our results provide a framework for targeting the host ubiquitin system in the development of host-based therapies for infectious disease. Compound C6 represents a promising tool with which to elucidate the role of DUBs in the macrophage response to infection.


Asunto(s)
Antivirales/farmacología , Animales , Enzimas Desubicuitinizantes/metabolismo , Humanos , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/metabolismo , Macrófagos/virología , Ratones , Norovirus/efectos de los fármacos , Norovirus/metabolismo , Replicación Viral/efectos de los fármacos
13.
Bioorg Med Chem ; 24(21): 5495-5504, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27647375

RESUMEN

The clinical selective estrogen receptor modulator tamoxifen is also a modest inhibitor of protein kinase C, a target implicated in several untreatable brain diseases such as amphetamine abuse. This inhibition and tamoxifen's ability to cross the blood brain barrier make it an attractive scaffold to conduct further SAR studies toward uncovering effective therapies for such diseases. Utilizing the known compound 6a as a starting template and guided by computational tools to derive physicochemical properties known to be important for CNS permeable drugs, the design and synthesis of a small series of novel triarylacrylonitrile analogues have been carried out providing compounds with enhanced potency and selectivity for PKC over the estrogen receptor relative to tamoxifen. Shortened synthetic routes compared to classical procedures have been developed for analogues incorporating a ß-phenyl ring, which involve installing dialkylaminoalkoxy side chains first off the α and/or α' rings of a precursor benzophenone and then condensing the resultant ketones with phenylacetonitrile anion. A second novel, efficient and versatile route utilizing Suzuki chemistry has also been developed, which will allow for the introduction of a wide range of ß-aryl or ß-heteroaryl moieties and side-chain substituents onto the acrylonitrile core. For analogues possessing a single side chain off the α- or α'-ring, novel 2D NMR experiments have been carried out that allow for unambiguous assignment of E- and Z-stereochemistry. From the SAR analysis, one compound, 6c, shows markedly increased potency and selectivity for inhibiting PKC with an IC50 of 80nM for inhibition of PKC protein substrate and >10µM for binding to the estrogen receptor α (tamoxifen IC50=20µM and 222nM, respectively). The data on 6c provide support for further exploration of PKC as a druggable target for the treatment of amphetamine abuse.


Asunto(s)
Acrilonitrilo/farmacología , Diseño de Fármacos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Tamoxifeno/farmacología , Acrilonitrilo/síntesis química , Acrilonitrilo/química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Tamoxifeno/química
14.
Mol Pharm ; 12(9): 3399-407, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26262434

RESUMEN

Understanding the mechanistic basis of prodrug delivery and activation is critical for establishing species-specific prodrug sensitivities necessary for evaluating preclinical animal models and potential drug-drug interactions. Despite significant adoption of prodrug methodologies for enhanced pharmacokinetics, functional annotation of prodrug activating enzymes is laborious and often unaddressed. Activity-based protein profiling (ABPP) describes an emerging chemoproteomic approach to assay active site occupancy within a mechanistically similar enzyme class in native proteomes. The serine hydrolase enzyme family is broadly reactive with reporter-linked fluorophosphonates, which have shown to provide a mechanism-based covalent labeling strategy to assay the activation state and active site occupancy of cellular serine amidases, esterases, and thioesterases. Here we describe a modified ABPP approach using direct substrate competition to identify activating enzymes for an ethyl ester prodrug, the influenza neuraminidase inhibitor oseltamivir. Substrate-competitive ABPP analysis identified carboxylesterase 1 (CES1) as an oseltamivir-activating enzyme in intestinal cell homogenates. Saturating concentrations of oseltamivir lead to a four-fold reduction in the observed rate constant for CES1 inactivation by fluorophosphonates. WWL50, a reported carbamate inhibitor of mouse CES1, blocked oseltamivir hydrolysis activity in human cell homogenates, confirming CES1 is the primary prodrug activating enzyme for oseltamivir in human liver and intestinal cell lines. The related carbamate inhibitor WWL79 inhibited mouse but not human CES1, providing a series of probes for analyzing prodrug activation mechanisms in different preclinical models. Overall, we present a substrate-competitive activity-based profiling approach for broadly surveying candidate prodrug hydrolyzing enzymes and outline the kinetic parameters for activating enzyme discovery, ester prodrug design, and preclinical development of ester prodrugs.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Inhibidores Enzimáticos/farmacología , Ésteres/farmacología , Oseltamivir/farmacología , Profármacos/farmacología , Animales , Línea Celular , Humanos , Hidrólisis , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Especificidad por Sustrato
15.
J Bacteriol ; 196(15): 2851-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24891441

RESUMEN

RecA is central to maintaining genome integrity in bacterial cells. Despite the near-ubiquitous conservation of RecA in eubacteria, the pathways that facilitate RecA loading and repair center assembly have remained poorly understood in Bacillus subtilis. Here, we show that RecA rapidly colocalizes with the DNA polymerase complex (replisome) immediately following DNA damage or damage-independent replication fork arrest. In Escherichia coli, the RecFOR and RecBCD pathways serve to load RecA and the choice between these two pathways depends on the type of damage under repair. We found in B. subtilis that the rapid localization of RecA to repair centers is strictly dependent on RecO and RecR in response to all types of damage examined, including a site-specific double-stranded break and damage-independent replication fork arrest. Furthermore, we provide evidence that, although RecF is not required for RecA repair center formation in vivo, RecF does increase the efficiency of repair center assembly, suggesting that RecF may influence the initial stages of RecA nucleation or filament extension. We further identify single-stranded DNA binding protein (SSB) as an additional component important for RecA repair center assembly. Truncation of the SSB C terminus impairs the ability of B. subtilis to form repair centers in response to damage and damage-independent fork arrest. With these results, we conclude that the SSB-dependent recruitment of RecOR to the replisome is necessary for loading and organizing RecA into repair centers in response to DNA damage and replication fork arrest.


Asunto(s)
Bacillus subtilis/genética , Proteínas Bacterianas/genética , Replicación del ADN/efectos de los fármacos , ADN Bacteriano/genética , Uracilo/análogos & derivados , Bacillus subtilis/citología , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Daño del ADN , Reparación del ADN , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Fluorescentes Verdes , Mutación , Rec A Recombinasas/genética , Rec A Recombinasas/metabolismo , Proteínas Recombinantes de Fusión , Uracilo/farmacología
16.
PLoS Pathog ; 8(7): e1002783, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792064

RESUMEN

Ubiquitin (Ub) is a vital regulatory component in various cellular processes, including cellular responses to viral infection. As obligate intracellular pathogens, viruses have the capacity to manipulate the ubiquitin (Ub) cycle to their advantage by encoding Ub-modifying proteins including deubiquitinases (DUBs). However, how cellular DUBs modulate specific viral infections, such as norovirus, is poorly understood. To examine the role of DUBs during norovirus infection, we used WP1130, a small molecule inhibitor of a subset of cellular DUBs. Replication of murine norovirus in murine macrophages and the human norovirus Norwalk virus in a replicon system were significantly inhibited by WP1130. Chemical proteomics identified the cellular DUB USP14 as a target of WP1130 in murine macrophages, and pharmacologic inhibition or siRNA-mediated knockdown of USP14 inhibited murine norovirus infection. USP14 is a proteasome-associated DUB that also binds to inositol-requiring enzyme 1 (IRE1), a critical mediator of the unfolded protein response (UPR). WP1130 treatment of murine macrophages did not alter proteasome activity but activated the X-box binding protein-1 (XBP-1) through an IRE1-dependent mechanism. In addition, WP1130 treatment or induction of the UPR also reduced infection of other RNA viruses including encephalomyocarditis virus, Sindbis virus, and La Crosse virus but not vesicular stomatitis virus. Pharmacologic inhibition of the IRE1 endonuclease activity partially rescued the antiviral effect of WP1130. Taken together, our studies support a model whereby induction of the UPR through cellular DUB inhibition blocks specific viral infections, and suggest that cellular DUBs and the UPR represent novel targets for future development of broad spectrum antiviral therapies.


Asunto(s)
Antivirales/farmacología , Nitrilos/farmacología , Norovirus/efectos de los fármacos , Piridinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/virología , Línea Celular , Línea Celular Tumoral , Cianoacrilatos , Proteínas de Unión al ADN/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/patogenicidad , Inhibidores Enzimáticos/farmacología , Humanos , Virus La Crosse/efectos de los fármacos , Virus La Crosse/patogenicidad , Macrófagos/virología , Proteínas de la Membrana/metabolismo , Ratones , Norovirus/fisiología , Virus Norwalk/efectos de los fármacos , Virus Norwalk/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción del Factor Regulador X , Virus Sindbis/efectos de los fármacos , Virus Sindbis/patogenicidad , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética , Replicación Viral/efectos de los fármacos , Proteína 1 de Unión a la X-Box
17.
Nat Chem Biol ; 8(3): 277-84, 2012 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-22286128

RESUMEN

Translocations involving the mixed lineage leukemia (MLL) gene result in human acute leukemias with very poor prognosis. The leukemogenic activity of MLL fusion proteins is critically dependent on their direct interaction with menin, a product of the multiple endocrine neoplasia (MEN1) gene. Here we present what are to our knowledge the first small-molecule inhibitors of the menin-MLL fusion protein interaction that specifically bind menin with nanomolar affinities. These compounds effectively reverse MLL fusion protein-mediated leukemic transformation by downregulating the expression of target genes required for MLL fusion protein oncogenic activity. They also selectively block proliferation and induce both apoptosis and differentiation of leukemia cells harboring MLL translocations. Identification of these compounds provides a new tool for better understanding MLL-mediated leukemogenesis and represents a new approach for studying the role of menin as an oncogenic cofactor of MLL fusion proteins. Our findings also highlight a new therapeutic strategy for aggressive leukemias with MLL rearrangements.


Asunto(s)
Antineoplásicos/farmacología , Leucemia/tratamiento farmacológico , Proteína de la Leucemia Mieloide-Linfoide/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Relación Estructura-Actividad
18.
Bioorg Med Chem ; 22(4): 1450-8, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24457091

RESUMEN

A series of degrasyn-like symmetrical compounds have been designed, synthesized, and screened against B cell malignancy (multiple myeloma, mantle cell lymphoma) cell lines. The lead compounds T5165804 and CP2005 showed higher nanomolar potency against these tumor cells in comparison to degrasyn and inhibited Usp9x activity in vitro and in intact cells. These observations suggest that this new class of compounds holds promise as cancer therapeutic agents.


Asunto(s)
Antineoplásicos/química , Nitrilos/química , Piridinas/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cianoacrilatos , Dimerización , Humanos , Modelos Moleculares , Mieloma Múltiple/tratamiento farmacológico , Nitrilos/farmacología , Nitrilos/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo
19.
Nucleic Acids Res ; 39(7): 2834-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21131277

RESUMEN

The enzyme tRNA-guanine transglycosylase (TGT) is involved in the queuosine modification of tRNAs in eukarya and eubacteria and in the archaeosine modification of tRNAs in archaea. However, the different classes of TGTs utilize different heterocyclic substrates (and tRNA in the case of archaea). Based on the X-ray structural analyses, an earlier study [Stengl et al. (2005) Mechanism and substrate specificity of tRNA-guanine transglycosylases (TGTs): tRNA-modifying enzymes from the three different kingdoms of life share a common catalytic mechanism. Chembiochem, 6, 1926-1939] has made a compelling case for the divergent evolution of the eubacterial and archaeal TGTs. The X-ray structure of the eukaryal class of TGTs is not known. We performed sequence homology and phylogenetic analyses, and carried out enzyme kinetics studies with the wild-type and mutant TGTs from Escherichia coli and human using various heterocyclic substrates that we synthesized. Observations with the Cys145Val (E. coli) and the corresponding Val161Cys (human) TGTs are consistent with the idea that the Cys145 evolved in eubacterial TGTs to recognize preQ(1) but not queuine, whereas the eukaryal equivalent, Val161, evolved for increased recognition of queuine and a concomitantly decreased recognition of preQ(1). Both the phylogenetic and kinetic analyses support the conclusion that all TGTs have divergently evolved to specifically recognize their cognate heterocyclic substrates.


Asunto(s)
Escherichia coli/enzimología , Evolución Molecular , Pentosiltransferasa/química , Secuencia de Aminoácidos , Guanina/análogos & derivados , Guanina/síntesis química , Guanina/química , Guanina/metabolismo , Humanos , Cinética , Datos de Secuencia Molecular , Mutación , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Filogenia , Pirimidinonas/síntesis química , Pirimidinonas/química , Pirimidinonas/metabolismo , Pirroles/síntesis química , Pirroles/química , Pirroles/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
20.
ACS Infect Dis ; 8(8): 1408-1421, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35772743

RESUMEN

Tuberculosis (TB) is one of the most significant world health problems, responsible for 1.5 M deaths in 2020, and yet, current treatments rely largely on 40 year old paradigms. Although the rifamycins (RIFs), best exemplified by the drug rifampin (RMP), represent a well-studied and therapeutically effective chemotype that targets the bacterial RNA polymerase (RNAP), these agents still suffer from serious drawbacks including the following: 3-9 month treatment times; cytochrome P450 (Cyp450) induction [particularly problematic for human immunodeficiency virus-Mycobacterium tuberculosis (MTB) co-infection]; and the existence of RIF-resistant (RIFR) MTB strains. We have utilized a structure-based drug design approach to synthesize and test 15 benzoxazinorifamycins (bxRIFs), congeners of the clinical candidate rifalazil, to minimize human pregnane X receptor (hPXR) activation while improving potency against MTB. We have determined the compounds' activation of the hPXR [responsible for inducing Cyp450 3A4 (CYP3A4)]. Compound IC50s have been determined against the wild-type and the most prevalent RIFR (ß-S450L) mutant MTB RNAPs. We have also determined their bactericidal activity against "normal" replicating MTB and a model for non-replicating, persister MTB. We have identified a minimal substitution and have probed larger substitutions that exhibit negligible hPXR activation (1.2-fold over the dimethyl sulfoxide control), many of which are 5- to 10-fold more potent against RNAPs and MTB than RMP. Importantly, we have analogues that are essentially equipotent against replicating MTB and non-replicating persister MTB, a property that is correlated with faster kill rates and may lead to shorter treatment durations. This work provides a proof of principle that the ansamycin core remains an attractive and effective scaffold for novel and dramatically improved RIFs.


Asunto(s)
Infecciones por VIH , Rifamicinas , Tuberculosis , Adulto , Infecciones por VIH/tratamiento farmacológico , Humanos , Receptor X de Pregnano , Rifampin/farmacología , Rifampin/uso terapéutico , Rifamicinas/farmacología , Rifamicinas/uso terapéutico , Tuberculosis/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA