Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34903667

RESUMEN

KRAS is mutated in 90% of human pancreatic ductal adenocarcinomas (PDACs). To function, KRAS must localize to the plasma membrane (PM) via a C-terminal membrane anchor that specifically engages phosphatidylserine (PtdSer). This anchor-binding specificity renders KRAS-PM localization and signaling capacity critically dependent on PM PtdSer content. We now show that the PtdSer lipid transport proteins, ORP5 and ORP8, which are essential for maintaining PM PtdSer levels and hence KRAS PM localization, are required for KRAS oncogenesis. Knockdown of either protein, separately or simultaneously, abrogated growth of KRAS-mutant but not KRAS-wild-type pancreatic cancer cell xenografts. ORP5 or ORP8 knockout also abrogated tumor growth in an immune-competent orthotopic pancreatic cancer mouse model. Analysis of human datasets revealed that all components of this PtdSer transport mechanism, including the PM-localized EFR3A-PI4KIIIα complex that generates phosphatidylinositol-4-phosphate (PI4P), and endoplasmic reticulum (ER)-localized SAC1 phosphatase that hydrolyzes counter transported PI4P, are significantly up-regulated in pancreatic tumors compared to normal tissue. Taken together, these results support targeting PI4KIIIα in KRAS-mutant cancers to deplete the PM-to-ER PI4P gradient, reducing PM PtdSer content. We therefore repurposed the US Food and Drug Administration-approved hepatitis C antiviral agent, simeprevir, as a PI4KIIIα inhibitor In a PDAC setting. Simeprevir potently mislocalized KRAS from the PM, reduced the clonogenic potential of pancreatic cancer cell lines in vitro, and abrogated the growth of KRAS-dependent tumors in vivo with enhanced efficacy when combined with MAPK and PI3K inhibitors. We conclude that the cellular ER-to-PM PtdSer transport mechanism is essential for KRAS PM localization and oncogenesis and is accessible to therapeutic intervention.


Asunto(s)
Antineoplásicos/farmacología , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Fosfatidilserinas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Receptores de Esteroides/metabolismo , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/genética , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Desnudos , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptores de Esteroides/genética , Simeprevir/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
ChemMedChem ; 16(23): 3548-3552, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34382337

RESUMEN

Over half a century since the description of the first antiviral drug, "old" re-emerging viruses and "new" emerging viruses still represent a serious threat to global health. Their high mutation rate and rapid selection of resistance toward common antiviral drugs, together with the increasing number of co-infections, make the war against viruses quite challenging. Herein we report a host-targeted approach, based on the inhibition of the lipid kinase PI4KIIIß, as a promising strategy for inhibiting the replication of multiple viruses hijacking this protein. We show that bithiazole inhibitors of PI4KIIIß block the replication of human rhinoviruses (hRV), Zika virus (ZIKV) and SARS-CoV-2 at low micromolar and sub-micromolar concentrations. However, while the anti-hRV/ZIKV activity can be directly linked to PI4KIIIß inhibition, the role of PI4KIIIß in SARS-CoV-2 entry/replication is debated.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antivirales/farmacología , Inhibidores Enzimáticos/química , Rhinovirus/fisiología , SARS-CoV-2/fisiología , Tiazoles/química , Replicación Viral/efectos de los fármacos , Virus Zika/fisiología , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Antivirales/química , Antivirales/metabolismo , COVID-19/patología , COVID-19/virología , Línea Celular , Estabilidad de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , SARS-CoV-2/aislamiento & purificación , Tiazoles/metabolismo , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/patología
3.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34155143

RESUMEN

A chromosome 1q21.3 region that is frequently amplified in diverse cancer types encodes phosphatidylinositol (PI)-4 kinase IIIß (PI4KIIIß), a key regulator of secretory vesicle biogenesis and trafficking. Chromosome 1q21.3-amplified lung adenocarcinoma (1q-LUAD) cells rely on PI4KIIIß for Golgi-resident PI-4-phosphate (PI4P) synthesis, prosurvival effector protein secretion, and cell viability. Here, we show that 1q-LUAD cells subjected to prolonged PI4KIIIß antagonist treatment acquire tolerance by activating an miR-218-5p-dependent competing endogenous RNA network that up-regulates PI4KIIα, which provides an alternative source of Golgi-resident PI4P that maintains prosurvival effector protein secretion and cell viability. These findings demonstrate an addiction to Golgi-resident PI4P synthesis in a genetically defined subset of cancers.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Cromosomas Humanos Par 1/genética , Amplificación de Genes , Aparato de Golgi/metabolismo , Fosfatos de Fosfatidilinositol/biosíntesis , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Fosfatos de Fosfatidilinositol/antagonistas & inhibidores , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/genética
4.
Adv Exp Med Biol ; 1274: 203-222, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32894512

RESUMEN

The lipid kinases that generate the lipid signalling phosphoinositides have been established as fundamental signalling enzymes that control numerous aspects of how cells respond to their extracellular environment. In addition, they play critical roles in regulating membrane trafficking and lipid transport within the cell. The class I phosphoinositide kinases which generate the critical lipid signal PIP3 are hyperactivated in numerous human pathologies including cancer, overgrowth syndromes, and primary immunodeficiencies. The type III phosphatidylinositol 4-kinase beta isoform (PI4KB), which are evolutionarily similar to the class I PI3Ks, have been found to be essential host factors mediating the replication of numerous devastating pathogenic viruses. Finally, targeting the parasite variant of PI4KB has been established as one of the most promising strategies for the development of anti-malarial and anti-cryptosporidium strategies. Therefore, the development of targeted isoform selective inhibitors for these enzymes are of paramount importance. The first generation of PI3K inhibitors have recently been clinically approved for a number of different cancers, highlighting their therapeutic value. This review will examine the history of the class I PI3Ks, and the type III PI4Ks, their relevance to human disease, and the structural basis for their regulation and inhibition by potent and selective inhibitors.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Enfermedades del Sistema Inmune/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Enfermedades Parasitarias/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Enfermedades de Inmunodeficiencia Primaria/tratamiento farmacológico , Virosis/tratamiento farmacológico , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Humanos , Enfermedades del Sistema Inmune/enzimología , Neoplasias/enzimología , Enfermedades Parasitarias/enzimología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Enfermedades de Inmunodeficiencia Primaria/enzimología , Virosis/enzimología
5.
mBio ; 11(2)2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127445

RESUMEN

Cryptosporidium parvum and Cryptosporidium hominis have emerged as major enteric pathogens of infants in the developing world, in addition to their known importance in immunocompromised adults. Although there has been recent progress in identifying new small molecules that inhibit Cryptosporidium sp. growth in vitro or in animal models, we lack information about their mechanism of action, potency across the life cycle, and cidal versus static activities. Here, we explored four potent classes of compounds that include inhibitors that likely target phosphatidylinositol 4 kinase (PI4K), phenylalanine-tRNA synthetase (PheRS), and several potent inhibitors with unknown mechanisms of action. We utilized monoclonal antibodies and gene expression probes for staging life cycle development to define the timing of when inhibitors were active during the life cycle of Cryptosporidium parvum grown in vitro These different classes of inhibitors targeted different stages of the life cycle, including compounds that blocked replication (PheRS inhibitors), prevented the segmentation of daughter cells and thus blocked egress (PI4K inhibitors), or affected sexual-stage development (a piperazine compound of unknown mechanism). Long-term cultivation of C. parvum in epithelial cell monolayers derived from intestinal stem cells was used to distinguish between cidal and static activities based on the ability of parasites to recover from treatment. Collectively, these approaches should aid in identifying mechanisms of action and for designing in vivo efficacy studies based on time-dependent concentrations needed to achieve cidal activity.IMPORTANCE Currently, nitazoxanide is the only FDA-approved treatment for cryptosporidiosis; unfortunately, it is ineffective in immunocompromised patients, has varied efficacy in immunocompetent individuals, and is not approved in infants under 1 year of age. Identifying new inhibitors for the treatment of cryptosporidiosis requires standardized and quantifiable in vitro assays for assessing potency, selectivity, timing of activity, and reversibility. Here, we provide new protocols for defining which stages of the life cycle are susceptible to four highly active compound classes that likely inhibit different targets in the parasite. We also utilize a newly developed long-term culture system to define assays for monitoring reversibility as a means of defining cidal activity as a function of concentration and time of treatment. These assays should provide valuable in vitro parameters to establish conditions for efficacious in vivo treatment.


Asunto(s)
Antiprotozoarios/farmacología , Cryptosporidium parvum/efectos de los fármacos , Cryptosporidium parvum/crecimiento & desarrollo , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Estadios del Ciclo de Vida/efectos de los fármacos , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antiprotozoarios/clasificación , Línea Celular , Línea Celular Tumoral , Inhibidores Enzimáticos/clasificación , Células Epiteliales/parasitología , Humanos
6.
Artículo en Inglés | MEDLINE | ID: mdl-31932368

RESUMEN

MMV390048 is a novel antimalarial compound that inhibits Plasmodium phosphatidylinositol-4-kinase. The safety, tolerability, pharmacokinetic profile, and antimalarial activity of MMV390048 were determined in healthy volunteers in three separate studies. A first-in-human, double-blind, randomized, placebo-controlled, single-ascending-dose study was performed. Additionally, a volunteer infection study investigated the antimalarial activity of MMV390048 using the Plasmodium falciparum induced blood-stage malaria (IBSM) model. Due to the high pharmacokinetic variability with the powder-in-bottle formulation used in both of these studies, a third study was undertaken to select a tablet formulation of MMV390048 to take forward into future studies. MMV390048 was generally well tolerated when administered as a single oral dose up to 120 mg, with rapid absorption and a long elimination half-life. Twelve adverse events were considered to be potentially related to MMV390048 in the first-in-human study but with no obvious correlation between these and MMV390048 dose or exposure. Although antimalarial activity was evident in the IBSM study, rapid recrudescence occurred in most subjects after treatment with 20 mg MMV390048, a dose expected to be subtherapeutic. Reformulation of MMV390048 into two tablet formulations (tartaric acid and Syloid) resulted in significantly reduced intersubject pharmacokinetic variability. Overall, the results of this study suggest that MMV390048 is well tolerated in humans, and the pharmacokinetic properties of the compound indicate that it has the potential to be used for antimalarial prophylaxis or inclusion in a single-dose cure. MMV390048 is currently being tested in a phase 2a study in Ethiopian adults with acute, uncomplicated falciparum or vivax malaria monoinfection. (The three clinical trials described here were each registered with ClinicalTrials.gov as follows: first-in-human study, registration no. NCT02230579; IBSM study, registration no. NCT02281344; and formulation optimization study, registration no. NCT02554799.).


Asunto(s)
Aminopiridinas/farmacología , Antimaláricos/farmacología , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Sulfonas/farmacología , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Adulto , Aminopiridinas/efectos adversos , Aminopiridinas/farmacocinética , Antimaláricos/efectos adversos , Antimaláricos/farmacocinética , Ensayos Clínicos Fase I como Asunto , Método Doble Ciego , Femenino , Voluntarios Sanos , Humanos , Masculino , Sulfonas/efectos adversos , Sulfonas/farmacocinética
7.
Eur J Med Chem ; 188: 112012, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31911293

RESUMEN

Starting from a bipyridine-sulfonamide scaffold, medicinal chemistry optimization leads to the discovery of a novel Plasmodium falciparum PI4K kinase (PfPI4K) inhibitor compound 15g (CHMFL-PI4K-127, IC50: 0.9 nM), which exhibits potent activity against 3D7 Plasmodium falciparum (P. falciparum) (EC50: 25.1 nM). CHMFL-PI4K-127 displays high selectivity against PfPI4K over human lipid and protein kinase. In addition, it exhibits EC50 values of 23-47 nM against a panel of the drug-resistant strains of P. falciparum. In vivo, the inhibitor demonstrates the favorable pharmacokinetic properties in both rats and mice. Furthermore, oral administration of CHMFL-PI4K-127 exhibits the antimalaria efficacy in both blood stage (80 mg/kg) and liver stage (1 mg/kg) of Plasmodium in infected rodent model. The results suggest that CHMFL-PI4K-127 might be a new potential drug candidate for malaria.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antimaláricos/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Hígado/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Piridinas/farmacología , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Antimaláricos/sangre , Antimaláricos/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/química , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/metabolismo , Piridinas/sangre , Piridinas/química , Relación Estructura-Actividad
8.
Phys Chem Chem Phys ; 21(39): 22103-22112, 2019 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-31570909

RESUMEN

Type III phosphatidylinositol 4 kinases (PI4KIIIs) are essential enzymes that are related to the replication of multiple RNA viruses. Understanding the interaction mechanisms of molecular compounds with the alpha and beta isoforms of PI4KIII (PI4KIIIα and PI4KIIIß) is of significance in the development of inhibitors that can bind to these two enzymes selectively. In this work, molecular dynamics (MD) simulations and binding free energy calculations were combined to investigate the binding modes of seven selected compounds to PI4KIIIα and PI4KIIIß. Analyses based on MD trajectories provide detailed interaction mechanisms of these compounds with PI4KIIIα and PI4KIIIß at the atomic level, and indicate that the selectivity of these compounds is mainly due to the structural difference of the binding pockets. It is expected that the detailed binding information found in this study can provide useful help for the structure-based design of selective inhibitors toward PI4KIIIα and PI4KIIIß.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/química , Secuencia de Aminoácidos , Dominio Catalítico , Concentración 50 Inhibidora , Estructura Molecular , Unión Proteica , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad , Termodinámica
9.
Comput Biol Chem ; 80: 79-89, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30928871

RESUMEN

The current study was set to discover selective Plasmodium falciparum phosphatidylinositol-4-OH kinase type III beta (pfPI4KB) inhibitors as potential antimalarial agents using combined structure-based and ligand-based drug discovery approach. A comparative model of pfPI4KB was first constructed and validated using molecular docking techniques. Performance of Autodock4.2 and Vina4 software in predicting the inhibitor-PI4KB binding mode and energy was assessed based on two Test Sets: Test Set I contained five ligands with resolved crystal structures with PI4KB, while Test Set II considered eleven compounds with known IC50 value towards PI4KB. The outperformance of Autodock as compared to Vina was reported, giving a correlation coefficient (R2) value of 0.87 and 0.90 for Test Set I and Test Set II, respectively. Pharmacophore-based screening was then conducted to identify drug-like molecules from ZINC database with physicochemical similarity to two potent pfPI4KB inhibitors -namely cpa and cpb. For each query inhibitor, the best 1000 hits in terms of TanimotoCombo scores were selected and subjected to molecular docking and molecular dynamics (MD) calculations. Binding energy was then estimated using molecular mechanics-generalized Born surface area (MM-GBSA) approach over 50 ns MD simulations of the inhibitor-pfPI4KB complexes. According to the calculated MM-GBSA binding energies, ZINC78988474 and ZINC20564116 were identified as potent pfPI4KB inhibitors with binding energies better than those of cpa and cpb, with ΔGbinding ≥ -34.56 kcal/mol. The inhibitor-pfPI4KB interaction and stability were examined over 50 ns MD simulation; as well the selectivity of the identified inhibitors towards pfPI4KB over PI4KB was reported.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Antimaláricos/metabolismo , Plasmodium falciparum/enzimología , Inhibidores de Proteínas Quinasas/metabolismo , 1-Fosfatidilinositol 4-Quinasa/química , Secuencia de Aminoácidos , Antimaláricos/química , Dominio Catalítico , Bases de Datos de Compuestos Químicos/estadística & datos numéricos , Descubrimiento de Drogas , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Alineación de Secuencia
10.
J Lipid Res ; 60(3): 683-693, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30626625

RESUMEN

The minor phospholipid, phosphatidylinositol 4-phosphate (PI4P), is emerging as a key regulator of lipid transfer in ER-membrane contact sites. Four different phosphatidylinositol 4-kinase (PI4K) enzymes generate PI4P in different membrane compartments supporting distinct cellular processes, many of which are crucial for the maintenance of cellular integrity but also hijacked by intracellular pathogens. While type III PI4Ks have been targeted by small molecular inhibitors, thus helping decipher their importance in cellular physiology, no inhibitors are available for the type II PI4Ks, which hinders investigations into their cellular functions. Here, we describe the identification of small molecular inhibitors of PI4K type II alpha (PI4K2A) by implementing a large scale small molecule high-throughput screening. A novel assay was developed that allows testing of selected inhibitors against PI4K2A in intact cells using a bioluminescence resonance energy transfer approach adapted to plate readers. The compounds disclosed here will pave the way to the optimization of PI4K2A inhibitors that can be used in cellular and animal studies to better understand the role of this enzyme in both normal and pathological states.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , 1-Fosfatidilinositol 4-Quinasa/química , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Transporte Biológico , Células COS , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Inhibidores Enzimáticos/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Conformación Proteica
11.
Autophagy ; 15(3): 466-477, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30290718

RESUMEN

Macroautophagy/autophagy plays an important role in the immune response to invasion by intracellular pathogens such as group A Streptococcus (GAS; Streptococcus pyogenes). We previously identified RAB30, a Golgi-resident GTPase, as a novel anti-bacterial autophagic regulator in the formation of GAS-containing autophagosome-like vacuoles (GcAVs); however, the precise mechanism underlying this process remains elusive. Here, we elucidate a novel property of RAB30: the ability to recruit PI4KB (phosphatidylinositol 4-kinase beta) to the Golgi apparatus and GcAVs. We found that trans-Golgi network (TGN) vesicles were incorporated into GcAVs via RAB30 to promote GcAV formation. Moreover, depletion of phosphatidylinositol-4-phosphate (PtdIns4P), a phosphatidylinositol enriched in the TGN, by wortmannin and phenylarsine oxide, followed by subsequent repletion with exogenous PtdIns4P revealed that PtdIns4P is crucial for GcAV formation. Furthermore, we identify an interaction between RAB30 and PI4KB, in which the knockdown of RAB30 decreased the localization of PI4KB to the TGN and GcAVs. Finally, PI4KB knockout suppressed autophagy by inhibiting GcAV formation, resulting in the increased survival of GAS. Our results demonstrate a novel autophagosomal formation mechanism involving coordinative functions of RAB30 and PI4KB distinct from those utilized in canonical autophagy. Abbreviations: GAS: group A Streptococcus; GcAVs: GAS-containing autophagosome-like vacuoles; PI4KB: phosphatidylinositol 4-kinase beta; PtdIns: phosphatidylinositol; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns4P: phosphatidylinositol-4-phosphate; PtdIns5P: phosphatidylinositol-5-phosphate; SLO: streptolysin O; TGN: trans-Golgi network; TGOLN2: trans-golgi network protein 2; PH: plekstrin homology; OSBP: oxysterol binding protein.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/metabolismo , Autofagosomas/microbiología , Aparato de Golgi/metabolismo , Streptococcus pyogenes/fisiología , Proteínas de Unión al GTP rab/metabolismo , Red trans-Golgi/metabolismo , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/genética , Autofagosomas/metabolismo , Autofagia/genética , Aparato de Golgi/microbiología , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Fosfatos de Fosfatidilinositol/química , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación , Vacuolas/metabolismo , Vacuolas/microbiología , Proteínas de Unión al GTP rab/antagonistas & inhibidores , Proteínas de Unión al GTP rab/genética , Red trans-Golgi/microbiología
12.
J Antimicrob Chemother ; 73(12): 3375-3384, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30219827

RESUMEN

Objectives: Many positive-stranded RNA viruses, including HCV, drastically remodel intracellular membranes to generate specialized environments for RNA replication. Phosphatidylinositol 4-kinase III (PI4KIII)α plays an essential role in the formation of HCV replication complexes and has therefore been explored as a potential drug target. Here, we characterized the anti-HCV activity of the PI4KIII inhibitors enviroxime and BF738735 and elucidated their mechanism of action. Methods: Antiviral assays were performed using HCV subgenomic replicons and infectious HCV. Enviroxime- and BF738735-resistant HCV replicons were generated by long-term culture with increasing compound concentrations. Intracellular localization of phosphatidylinositol 4-phosphate (PI4P) lipids was analysed by confocal microscopy. Results: HCV subgenomic replicons resistant to either enviroxime or BF738735 proved cross-resistant and carried mutations in the NS3, NS4B and NS5A genes. Knockdown of PI4KIIIß by small interfering RNA (siRNA) did not affect the replication of the HCV subgenomic replicon in this study. Furthermore, the compounds did not affect PI4P lipid levels at the replication complexes nor the phosphorylation status of NS5A, activities attributed to PI4KIIIα. Interestingly, the broad-spectrum phosphoinositide 3-kinase (PI3K) inhibitor LY294002 proved to be 10-fold less effective against the resistant replicons. In addition, enviroxime and BF738735 inhibited several PI3Ks in enzymatic assays. Conclusions: Contrary to assumptions, our data indicate that PI4KIIIα and PI4KIIIß are not the main targets for the anti-HCV activity of enviroxime and BF738735. Instead, we demonstrated that both molecules impede HCV replication at least partially by an inhibitory effect on PI3Ks. Moreover, HCV is able to bypass PI3K inhibition by acquiring mutations in its genome.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antivirales/farmacología , Bencimidazoles/farmacología , Hepacivirus/crecimiento & desarrollo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Replicación Viral/efectos de los fármacos , Línea Celular , Análisis Mutacional de ADN , Farmacorresistencia Viral , Hepatocitos/enzimología , Hepatocitos/virología , Humanos , Oximas , Pase Seriado , Sulfonamidas , Proteínas no Estructurales Virales/genética
13.
Artículo en Inglés | MEDLINE | ID: mdl-30249687

RESUMEN

The in vivo antimalarial efficacies of two phosphatidylinositol 4-kinase (PI4K) inhibitors, a 3,5-diaryl-2-aminopyrazine sulfoxide and its corresponding sulfone metabolite, were evaluated in the NOD-scid IL2Rγnull (NSG) murine malaria disease model of Plasmodium falciparum infection. We hypothesized that the sulfoxide would serve as a more soluble prodrug for the sulfone, which would lead to improved drug exposure with oral dosing. Both compounds had similar efficacy (90% effective dose [ED90], 0.1 mg kg-1 of body weight) across a quadruple-dose regimen. Pharmacokinetic profiling revealed rapid sulfoxide clearance via conversion to sulfone, with sulfone identified as the major active metabolite. When the sulfoxide was dosed, the exposure of the sulfone achieved was as much as 2.9-fold higher than when the sulfone was directly dosed, thereby demonstrating that the sulfoxide served as an effective prodrug for the treatment of malaria.


Asunto(s)
Antimaláricos/farmacología , Malaria Falciparum/tratamiento farmacológico , Parasitemia/tratamiento farmacológico , Profármacos/farmacología , Pirazinas/farmacología , Sulfonas/farmacología , Sulfóxidos/farmacología , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/genética , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Antimaláricos/sangre , Antimaláricos/síntesis química , Antimaláricos/farmacocinética , Biotransformación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Eritrocitos/parasitología , Expresión Génica , Humanos , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Parasitemia/patología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Plasmodium falciparum/crecimiento & desarrollo , Profármacos/síntesis química , Profármacos/farmacocinética , Pirazinas/sangre , Pirazinas/síntesis química , Pirazinas/farmacocinética , Sulfonas/sangre , Sulfonas/síntesis química , Sulfonas/farmacocinética , Sulfóxidos/sangre , Sulfóxidos/síntesis química , Sulfóxidos/farmacocinética , Resultado del Tratamiento
14.
J Med Chem ; 61(13): 5692-5703, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29889526

RESUMEN

A novel 2,8-disubstituted-1,5-naphthyridine hit compound stemming from the open access Medicines for Malaria Venture Pathogen Box formed a basis for a hit-to-lead medicinal chemistry program. Structure-activity relationship investigations resulted in compounds with potent antiplasmodial activity against both chloroquine sensitive (NF54) and multidrug resistant (K1) strains of the human malaria parasite Plasmodium falciparum. In the humanized P. falciparum mouse efficacy model, one of the frontrunner compounds showed in vivo efficacy at an oral dose of 4 × 50 mg·kg-1. In vitro mode-of-action studies revealed Plasmodium falciparum phosphatidylinositol-4-kinase as the target.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Malaria/tratamiento farmacológico , Naftiridinas/química , Naftiridinas/farmacología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , 1-Fosfatidilinositol 4-Quinasa/química , Animales , Antimaláricos/química , Antimaláricos/farmacocinética , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Modelos Animales de Enfermedad , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratones , Modelos Moleculares , Naftiridinas/farmacocinética , Naftiridinas/uso terapéutico , Plasmodium falciparum/fisiología , Conformación Proteica , Relación Estructura-Actividad , Distribución Tisular
15.
J Biol Chem ; 292(48): 19743-19751, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28972151

RESUMEN

It is well known that the reactive oxygen species NO can trigger cell death in plants and other organisms, but the underlying molecular mechanisms are not well understood. Here we provide evidence that NO may trigger cell death in tomato (Solanum lycopersicum) by inhibiting the activity of phosphoinositide-dependent kinase 1 (SlPDK1), a conserved negative regulator of cell death in yeasts, mammals, and plants, via S-nitrosylation. Biotin-switch assays indicated that SlPDK1 is a target of S-nitrosylation. Moreover, the kinase activity of SlPDK1 was inhibited by S-nitrosoglutathione in a concentration-dependent manner, indicating that SlPDK1 activity is abrogated by S-nitrosylation. The S-nitrosoglutathione-induced inhibition was reversible in the presence of a reducing agent but additively enhanced by hydrogen peroxide (H2O2). Our LC-MS/MS analyses further indicated that SlPDK1 is primarily S-nitrosylated on a cysteine residue at position 128 (Cys128), and substitution of Cys128 with serine completely abolished SlPDK1 kinase activity, suggesting that S-nitrosylation of Cys128 is responsible for SlPDK1 inhibition. In summary, our results establish a potential link between NO-triggered cell death and inhibition of the kinase activity of tomato PDK1.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Inhibidores de Proteínas Quinasas/farmacología , S-Nitrosoglutatión/farmacología , Solanum lycopersicum/enzimología , Aldehído Oxidorreductasas/genética , Muerte Celular , Cromatografía Liquida , Cisteína/metabolismo , Silenciador del Gen , Solanum lycopersicum/citología , Solanum lycopersicum/genética , Espectrometría de Masas en Tándem
16.
Nature ; 546(7658): 376-380, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28562588

RESUMEN

Diarrhoeal disease is responsible for 8.6% of global child mortality. Recent epidemiological studies found the protozoan parasite Cryptosporidium to be a leading cause of paediatric diarrhoea, with particularly grave impact on infants and immunocompromised individuals. There is neither a vaccine nor an effective treatment. Here we establish a drug discovery process built on scalable phenotypic assays and mouse models that take advantage of transgenic parasites. Screening a library of compounds with anti-parasitic activity, we identify pyrazolopyridines as inhibitors of Cryptosporidium parvum and Cryptosporidium hominis. Oral treatment with the pyrazolopyridine KDU731 results in a potent reduction in intestinal infection of immunocompromised mice. Treatment also leads to rapid resolution of diarrhoea and dehydration in neonatal calves, a clinical model of cryptosporidiosis that closely resembles human infection. Our results suggest that the Cryptosporidium lipid kinase PI(4)K (phosphatidylinositol-4-OH kinase) is a target for pyrazolopyridines and that KDU731 warrants further preclinical evaluation as a drug candidate for the treatment of cryptosporidiosis.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Criptosporidiosis/tratamiento farmacológico , Criptosporidiosis/parasitología , Cryptosporidium/efectos de los fármacos , Cryptosporidium/enzimología , Pirazoles/farmacología , Piridinas/farmacología , Animales , Animales Recién Nacidos , Bovinos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Huésped Inmunocomprometido , Interferón gamma/deficiencia , Interferón gamma/genética , Masculino , Ratones , Ratones Noqueados , Pirazoles/química , Pirazoles/farmacocinética , Piridinas/química , Piridinas/farmacocinética , Ratas , Ratas Wistar
17.
Sci Transl Med ; 9(387)2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28446690

RESUMEN

As part of the global effort toward malaria eradication, phenotypic whole-cell screening revealed the 2-aminopyridine class of small molecules as a good starting point to develop new antimalarial drugs. Stemming from this series, we found that the derivative, MMV390048, lacked cross-resistance with current drugs used to treat malaria. This compound was efficacious against all Plasmodium life cycle stages, apart from late hypnozoites in the liver. Efficacy was shown in the humanized Plasmodium falciparum mouse model, and modest reductions in mouse-to-mouse transmission were achieved in the Plasmodium berghei mouse model. Experiments in monkeys revealed the ability of MMV390048 to be used for full chemoprotection. Although MMV390048 was not able to eliminate liver hypnozoites, it delayed relapse in a Plasmodium cynomolgi monkey model. Both genomic and chemoproteomic studies identified a kinase of the Plasmodium parasite, phosphatidylinositol 4-kinase, as the molecular target of MMV390048. The ability of MMV390048 to block all life cycle stages of the malaria parasite suggests that this compound should be further developed and may contribute to malaria control and eradication as part of a single-dose combination treatment.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Aminopiridinas/uso terapéutico , Antimaláricos/uso terapéutico , Sulfonas/uso terapéutico , Aminopiridinas/farmacología , Animales , Antimaláricos/farmacología , Femenino , Malaria/tratamiento farmacológico , Malaria/enzimología , Masculino , Ratones , Ratones SCID , Pruebas de Sensibilidad Parasitaria , Plasmodium/efectos de los fármacos , Plasmodium/patogenicidad , Sulfonas/farmacología
18.
J Med Chem ; 60(1): 100-118, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28004945

RESUMEN

Phosphatidylinositol 4-kinase IIIß (PI4KB) is indispensable for the replication of various positive-sense single stranded RNA viruses, which hijack this cellular enzyme to remodel intracellular membranes of infected cells to set up the functional replication machinery. Therefore, the inhibition of this PI4K isoform leads to the arrest of viral replication. Here, we report on the synthesis of novel PI4KB inhibitors, which were rationally designed based on two distinct structural types of inhibitors that bind in the ATP binding side of PI4KB. These "hybrids" not only excel in outstanding inhibitory activity but also show high selectivity to PI4KB compared to other kinases. Thus, these compounds exert selective nanomolar or even subnanomolar activity against PI4KB as well as profound antiviral effect against hepatitis C virus, human rhinovirus, and coxsackievirus B3. Our crystallographic analysis unveiled the exact position of the side chains and explains their extensive contribution to the inhibitory activity.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antivirales/química , Antivirales/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Diseño de Fármacos , Células HeLa , Humanos , Estructura Molecular
19.
Int J Radiat Oncol Biol Phys ; 96(4): 867-876, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27788957

RESUMEN

PURPOSE: To investigate which isotype of phosphatidylinositol 4-kinase (PI4K) may affect radiosensitivity and examine whether anti-hepatitis C viral (HCV) agents, some of which have been shown to inhibit PI4K IIIα activity, could be repositioned as a radiosensitizer in human cancer cells. METHODS AND MATERIALS: U251, BT474, and HepG2 cell lines and normal human astrocyte were used. Ribonucleic acid interference, clonogenic assays, Western blotting, immunofluorescence, annexin V assay, lysotracker staining, and ß-galactosidase assay were performed. RESULTS: Of the 4 PI4K isotypes, specific inhibition of IIIα increased radiosensitivity. For pharmacologic inhibition of PI4K IIIα, we screened 9 anti-HCV agents by half-maximal inhibitory concentration assay. Simeprevir was selected, and its inhibition of PI4K IIIα activity was confirmed. Combination of simeprevir treatment and radiation significantly attenuated expression of phospho-phospho-PKC and phospho-Akt and increased radiation-induced cell death in tested cell lines. Pretreatment with simeprevir prolonged γH2AX foci formation and down-regulation of phospho-DNA-PKcs, indicating impairment of nonhomologous end-joining repair. Cells pretreated with simeprevir exhibited mixed modes of cell death, including apoptosis and autophagy. CONCLUSION: These data demonstrate that targeting PI4K IIIα using an anti-HCV agent is a viable approach to enhance the therapeutic efficacy of radiation therapy in various human cancers, such as glioma, breast, and hepatocellular carcinoma.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antivirales/uso terapéutico , Reparación del ADN/efectos de los fármacos , Reposicionamiento de Medicamentos , Inhibidores de Proteasas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Simeprevir/uso terapéutico , Apoptosis , Astrocitos/efectos de los fármacos , Astrocitos/efectos de la radiación , Autofagia , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Regulación hacia Abajo , Activación Enzimática , Femenino , Glioma/tratamiento farmacológico , Glioma/radioterapia , Histonas/efectos de los fármacos , Humanos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño , Ensayo de Tumor de Célula Madre , beta-Galactosidasa/análisis
20.
Antimicrob Agents Chemother ; 60(10): 6402-6, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27480860

RESUMEN

Encephalomyocarditis virus (EMCV), like hepatitis C virus (HCV), requires phosphatidylinositol 4-kinase IIIα (PI4KA) for genome replication. Here, we demonstrate that tyrphostin AG1478, a known epidermal growth factor receptor (EGFR) inhibitor, also inhibits PI4KA activity, both in vitro and in cells. AG1478 impaired replication of EMCV and HCV but not that of an EMCV mutant previously shown to escape PI4KA inhibition. This work uncovers novel cellular and antiviral properties of AG1478, a compound previously regarded only as a cancer chemotherapy agent.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antivirales/farmacología , Virus de la Encefalomiocarditis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Quinazolinas/farmacología , Tirfostinos/farmacología , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Relación Dosis-Respuesta a Droga , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/fisiología , Células HeLa/efectos de los fármacos , Células HeLa/virología , Hepacivirus/fisiología , Humanos , Terapia Molecular Dirigida/métodos , Mutación , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA