Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Chem Biodivers ; 20(6): e202300410, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37088929

RESUMEN

Two new naphthyridine compounds, 4-methoxycarbonyl-5-oxo-1,6-naphthyridine (1) and 5-methoxycarbonyl-4-oxo-1,6-naphthyridine (2) were obtained from the MeOH extracts of sponge Aaptos suberitoides. Their structures were determined by spectroscopic methods, including HR-ESI-MS, 1D-NMR (1 H-NMR, 13 C-NMR), 2D-NMR (COSY, HSQC, HMBC). The structure of compound 1 was further confirmed via single crystal X-ray diffraction analysis. Compound 1 was found to reduce NO production in LPS-induced RAW 264.7 macrophages with IC50 value of 0.15 mM. In addition, it decreased the mRNA expression levels of pro-inflammatory mediators, such as the tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1ß (IL-1ß), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) in LPS-induced macrophages. It also decreased the protein expression of iNOS and COX-2 in LPS-induced macrophages. Mechanistic studies further revealed that compound 1 inhibited the mitogen-activated protein kinase (MAPK), and activated the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) signaling pathways in LPS-induced RAW 264.7 macrophages.


Asunto(s)
Lipopolisacáridos , Proteínas Quinasas Activadas por Mitógenos , Animales , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/farmacología , Lipopolisacáridos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Células RAW 264.7 , Antiinflamatorios/farmacología , Antiinflamatorios/metabolismo , Transducción de Señal , Macrófagos , Naftiridinas/farmacología , Naftiridinas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Hemo-Oxigenasa 1/metabolismo , Óxido Nítrico/metabolismo
2.
Bioorg Med Chem Lett ; 49: 128314, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34391891

RESUMEN

A series of IDO1 inhibitors containing a decahydroquinoline, decahydro-1,6-naphthyridine, or octahydro-1H-pyrrolo[3,2-c]pyridine scaffold were identified with good cellular and human whole blood activity against IDO1. These inhibitors contain multiple chiral centers and all diastereomers were separated. The absolute stereochemistry of each isomers were not determined. Compounds 15 and 27 stood out as leads due to their good cellular as well as human whole blood IDO1 inhibition activity, low unbound clearance, and reasonable mean residence time in rat cassette PK studies.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Naftiridinas/farmacología , Pirroles/farmacología , Quinolinas/farmacología , Animales , Dominio Catalítico , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Células HeLa , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Simulación del Acoplamiento Molecular , Naftiridinas/síntesis química , Naftiridinas/metabolismo , Naftiridinas/farmacocinética , Pirroles/síntesis química , Pirroles/metabolismo , Pirroles/farmacocinética , Quinolinas/síntesis química , Quinolinas/metabolismo , Quinolinas/farmacocinética , Ratas , Estereoisomerismo , Relación Estructura-Actividad
3.
Proc Natl Acad Sci U S A ; 115(44): 11232-11237, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30327344

RESUMEN

Understanding how antibiotic-producing bacteria deal with highly reactive chemicals will ultimately guide therapeutic strategies to combat the increasing clinical resistance crisis. Here, we uncovered a distinctive self-defense strategy featured by a secreted oxidoreductase NapU to perform extracellularly oxidative activation and conditionally overoxidative inactivation of a matured prodrug in naphthyridinomycin (NDM) biosynthesis from Streptomyces lusitanus NRRL 8034. It was suggested that formation of NDM first involves a nonribosomal peptide synthetase assembly line to generate a prodrug. After exclusion and prodrug maturation, we identified a pharmacophore-inactivated intermediate, which required reactivation by NapU via oxidative C-H bond functionalization extracellularly to afford NDM. Beyond that, NapU could further oxidatively inactivate the NDM pharmacophore to avoid self-cytotoxicity if they coexist longer than necessary. This discovery represents an amalgamation of sophisticatedly temporal and spatial shielding mode conferring self-resistance in antibiotic biosynthesis from Gram-positive bacteria.


Asunto(s)
Antibacterianos/metabolismo , Profármacos/metabolismo , Streptomyces/metabolismo , Naftiridinas/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , Péptido Sintasas/metabolismo
4.
J Biol Chem ; 294(37): 13545-13559, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31341017

RESUMEN

The homeodomain-interacting protein kinase (HIPK) family is comprised of four nuclear protein kinases, HIPK1-4. HIPK proteins phosphorylate a diverse range of transcription factors involved in cell proliferation, differentiation, and apoptosis. HIPK2, thus far the best-characterized member of this largely understudied family of protein kinases, plays a role in the activation of p53 in response to DNA damage. Despite this tumor-suppressor function, HIPK2 is also found overexpressed in several cancers, and its hyperactivation causes chronic fibrosis. There are currently no structures of HIPK2 or of any other HIPK kinase. Here, we report the crystal structure of HIPK2's kinase domain bound to CX-4945, a casein kinase 2α (CK2α) inhibitor currently in clinical trials against several cancers. The structure, determined at 2.2 Å resolution, revealed that CX-4945 engages the HIPK2 active site in a hybrid binding mode between that seen in structures of CK2α and Pim1 kinases. The HIPK2 kinase domain crystallized in the active conformation, which was stabilized by phosphorylation of the activation loop. We noted that the overall kinase domain fold of HIPK2 closely resembles that of evolutionarily related dual-specificity tyrosine-regulated kinases (DYRKs). Most significant structural differences between HIPK2 and DYRKs included an absence of the regulatory N-terminal domain and a unique conformation of the CMGC-insert region and of a newly defined insert segment in the αC-ß4 loop. This first crystal structure of HIPK2 paves the way for characterizing the understudied members of the HIPK family and for developing HIPK2-directed therapies for managing cancer and fibrosis.


Asunto(s)
Proteínas Portadoras/química , Proteínas Serina-Treonina Quinasas/química , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Portadoras/clasificación , Proteínas Portadoras/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Evolución Molecular , Humanos , Simulación de Dinámica Molecular , Naftiridinas/química , Naftiridinas/metabolismo , Fenazinas , Filogenia , Unión Proteica , Proteínas Serina-Treonina Quinasas/clasificación , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia
5.
Alcohol Clin Exp Res ; 44(2): 354-367, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31840823

RESUMEN

Alcohol use disorder (AUD) is a chronic, relapsing disorder that is characterized by the compulsive use of alcohol despite numerous health, social, and economic consequences. Initially, the use of alcohol is driven by positive reinforcement. Over time, however, alcohol use can take on a compulsive quality that is driven by the desire to avoid the negative consequences of abstinence, including negative affect and heightened stress/anxiety. This transition from positive reinforcement- to negative reinforcement-driven consumption involves the corticotropin-releasing factor (CRF) system, although mounting evidence now suggests that the CRF system interacts with other neural systems to ultimately produce behaviors that are symptomatic of compulsive alcohol use, such as the hypocretin (Hcrt) system. Hypocretins are produced exclusively in the hypothalamus, but Hcrt neurons project widely throughout the brain and reach regions that perform regulatory functions for numerous behavioral and physiological responses-including the infralimbic cortex (IL) of the medial prefrontal cortex (mPFC). Although the entire mPFC undergoes neuroadaptive changes following prolonged alcohol exposure, the IL appears to undergo more robust changes compared with other mPFC substructures. Evidence to date suggests that the IL is likely involved in EtOH-seeking behavior, but ambiguities with respect to the specific role of the IL in this regard make it difficult to draw definitive conclusions. Furthermore, the manner in which CRF interacts with Hcrt in this region as it pertains to alcohol-seeking behavior is largely unknown, although immunohistochemical and electrophysiological experiments have shown that CRF and Hcrt directly interact in the mPFC, suggesting that the interaction between CRF and Hcrt in the IL may be critically important for the development and subsequent maintenance of compulsive alcohol seeking. This review aims to consolidate recent literature regarding the role of the IL in alcohol-seeking behavior and to discuss evidence that supports a functional interaction between Hcrt and CRF in the IL.


Asunto(s)
Alcoholismo/metabolismo , Conducta Compulsiva/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Comportamiento de Búsqueda de Drogas/fisiología , Orexinas/metabolismo , Corteza Prefrontal/metabolismo , Alcoholismo/tratamiento farmacológico , Animales , Benzoxazoles/metabolismo , Benzoxazoles/farmacología , Benzoxazoles/uso terapéutico , Conducta Compulsiva/tratamiento farmacológico , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Humanos , Naftiridinas/metabolismo , Naftiridinas/farmacología , Naftiridinas/uso terapéutico , Corteza Prefrontal/efectos de los fármacos , Unión Proteica/fisiología , Urea/análogos & derivados , Urea/metabolismo , Urea/farmacología , Urea/uso terapéutico
6.
Bioorg Med Chem Lett ; 30(23): 127517, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32911078

RESUMEN

A series of Torin2, a second-generation ATP-competitive inhibitor, analogues were biologically characterized to identify their potential for ATR and mTOR kinase inhibition. Compound SPK 98 was observed to inhibit ATR/mTOR kinase selectively over ATM kinase in HCT 116 cell line. In addition to that, SPK 98 on 30 min incubation with human, mice and rat liver microsomes showed improved properties with an increased half-life (a maximum T ½ of 157 min) and internal clearance in mouse as compared to Torin2. Further, SPK 98 was also noticed to indulge in inducing premature chromatin condensation as a result of ATR/mTOR kinase inhibition at 50 nM. In a nutshell, our work presents the identification and characterization of SPK 98, a small molecule inhibitor, which exhibits improved specific inhibition for ATR at a lower concentration than Torin2.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Naftiridinas/farmacología , Fármacos Fotosensibilizantes/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antineoplásicos/metabolismo , Cromatina/metabolismo , ADN/efectos de la radiación , Daño del ADN/efectos de la radiación , Células HCT116 , Humanos , Ratones , Microsomas Hepáticos/metabolismo , Naftiridinas/metabolismo , Fármacos Fotosensibilizantes/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Estabilidad Proteica , Ratas , Transducción de Señal/efectos de los fármacos , Rayos Ultravioleta
7.
Bioorg Med Chem ; 28(12): 115555, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32503697

RESUMEN

New N-substituted-3-phenyl-1,6-naphthyridinone derivatives are designed and synthesized, based on structural modification of our previously reported compound 3. Extensive enzyme-based SAR studies and PK evaluation led to the discovery of compound 4r, with comparable c-Met potency to that of Cabozantinib and high VEGFR-2 selectivity, while Cabozantinib displayed no VEGFR-2 selectivity. More importantly, at oral doses of 45 mg/kg (Q.D.), compound 4r exhibits significant tumor growth inhibition (93%) in a U-87MG human gliobastoma xenograft model. The promising selectivity against VEGFR-2 and excellent tumor growth inhibition of compound 4r suggest that it could be used as a new lead molecule for further discovery of selective type II c-Met inhibitors.


Asunto(s)
Diseño de Fármacos , Naftiridinas/química , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Quinolinas/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Simulación del Acoplamiento Molecular , Naftiridinas/metabolismo , Naftiridinas/farmacología , Naftiridinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/metabolismo , Relación Estructura-Actividad , Trasplante Heterólogo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Bioorg Med Chem ; 28(24): 115819, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33120078

RESUMEN

The exploitation of GLU988 and LYS903 residues in PARP1 as targets to design isoquinolinone (I & II) and naphthyridinone (III) analogues is described. Compounds of structure I have good biochemical and cellular potency but suffered from inferior PK. Constraining the linear propylene linker of structure I into a cyclopentene ring (II) offered improved PK parameters, while maintaining potency for PARP1. Finally, to avoid potential issues that may arise from the presence of an anilinic moiety, the nitrogen substituent on the isoquinolinone ring was incorporated as part of the bicyclic ring. This afforded a naphthyridinone scaffold, as shown in structure III. Further optimization of naphthyridinone series led to identification of a novel and highly potent PARP1 inhibitor 34, which was further characterized as preclinical candidate molecule. Compound 34 is orally bioavailable and displayed favorable pharmacokinetic (PK) properties. Compound 34 demonstrated remarkable antitumor efficacy both as a single-agent as well as in combination with chemotherapeutic agents in the BRCA1 mutant MDA-MB-436 breast cancer xenograft model. Additionally, compound 34 also potentiated the effect of agents such as temozolomide in breast cancer, pancreatic cancer and Ewing's sarcoma models.


Asunto(s)
Antineoplásicos/química , Naftiridinas/química , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Quinolonas/química , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sitios de Unión , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Semivida , Humanos , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Naftiridinas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Quinolonas/metabolismo , Relación Estructura-Actividad , Trasplante Heterólogo
9.
Chembiochem ; 20(23): 2903-2910, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31087756

RESUMEN

We introduce the concept of molecular glues for RNA, in which specific RNA-binding small molecules induce designed structural changes in target functional RNAs, resulting in modulation of the functions. (Z)-NCTS is an RNA-mismatch-binding small molecule that recognizes 5'-r(XGG)-3'/5'-r(XGG)-3' sequences (X=U or A) and acts as a molecular glue for RNA. The binding of (Z)-NCTS brings two distinct 5'-r(XGG)-3' domains into contact with each other, and this can result in higher-order structural changes of target RNAs. We applied (Z)-NCTS to induce the formation of a proposed tertiary structure of a ribozyme together with activation of RNA-cleaving ability. The concept of a molecular glue could inspire new small-molecule-based strategies for regulating biological functions: a synthetic small molecule targeting functional RNAs could regulate the RNA structure and function.


Asunto(s)
Naftiridinas/farmacología , Conformación de Ácido Nucleico/efectos de los fármacos , ARN Catalítico/metabolismo , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Secuencias Invertidas Repetidas , Naftiridinas/metabolismo , Hibridación de Ácido Nucleico/efectos de los fármacos , ARN Catalítico/genética
10.
Arterioscler Thromb Vasc Biol ; 38(9): 2217-2224, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30026269

RESUMEN

Objective- Drug-eluting stents eluting canonical mTOR (mammalian target of rapamycin) inhibitors are widely used to treat coronary artery disease but accelerate the development of atherosclerosis within the stent (neoatherosclerosis)-a leading cause of late stent failure. We recently showed that canonical mTOR inhibitors bind FKBP12.6 (12.6-kDa FK506-binding protein 12), displace it from calcium release channels, resulting in activation of PKCα (protein kinase Cα) and dissociation of p-120-catenin (p120) from VE-CAD (vascular endothelial cadherin; promoting endothelial barrier dysfunction [EBD]). However, the relevance of these findings to drug-eluting stents remains unknown. Newer generation direct mTOR kinase inhibitors do not bind FKBP12.6 and offer the potential of improving endothelial barrier function while maintaining antirestenotic efficacy, but their actual effects are unknown. We examined the effects of 2 different pharmacological targeting strategies-canonical mTOR inhibitor everolimus and mTOR kinase inhibitors Torin-2-on EBD after stenting. Approach and Results- Using the rabbit model of stenting and a combination of Evans blue dye, confocal and scanning electron microscopy studies, everolimus-eluting stents resulted in long-term EBD compared with bare metal stents. EBD was mitigated by using stents that eluted mTOR kinase inhibitors (Torin-2-eluting stent). At 60 days after stent placement, everolimus-eluting stents demonstrated large areas of Evans blue dye staining and evidence of p120 VE-CAD dissociation consistent with EBD. These findings were absent in bare metal stents and significantly attenuated in Torin-2-eluting stent. As proof of concept of the role of EBD in neoatherosclerosis, 100 days after stenting, animals were fed an enriched cholesterol diet for an additional 30 days. Everolimus-eluting stents demonstrated significantly more macrophage infiltration (consistent with neoatherosclerosis) compared with both bare metal stents and Torin-2-eluting stent. Conclusions- Our results pinpoint interactions between FKBP12.6 and canonical mTOR inhibitors as a major cause of vascular permeability and neoatherosclerosis, which can be overcome by using mTOR kinase inhibitors. Our study suggests further refinement of molecular targeting of the mTOR complex may be a promising strategy (Graphic Abstract).


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Stents Liberadores de Fármacos , Endotelio Vascular/metabolismo , Everolimus/farmacología , Naftiridinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Cateninas/metabolismo , Activación Enzimática , Everolimus/metabolismo , Masculino , Modelos Animales , Naftiridinas/metabolismo , Prueba de Estudio Conceptual , Proteína Quinasa C-alfa/metabolismo , Conejos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Catenina delta
11.
Bioorg Med Chem ; 27(10): 2140-2148, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30952388

RESUMEN

Small-molecule modulators, along with antisense oligonucleotide, would be powerful tools and potential drug candidates for modulating miRNA-related gene expressions. The mechanism of the inhibitory effect of the C-bulge binding small molecule BzDANP for the Dicer processing reaction of pre-miR-136 was discussed on the data obtained by SPR, NMR, and kinetic analysis for Dicer processing. SPR and NMR analysis showed the preference of BzDANP binding to the C-bulge. Michaelis-Menten analysis suggested the formation of a ternary complex pre-miR-136-BzDANP-Dicer during the Dicer-cleavage reaction of pre-miR-136 in the presence of BzDANP. The inhibitory effect of BzDANP is likely attributed to the slower reaction from the ternary complex than that from the binary pre-miR-136-Dicer complex.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , MicroARNs/química , Naftiridinas/química , Ribonucleasa III/metabolismo , Bibliotecas de Moléculas Pequeñas/química , ARN Helicasas DEAD-box/antagonistas & inhibidores , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Espectroscopía de Resonancia Magnética , MicroARNs/genética , MicroARNs/metabolismo , Mutación , Naftiridinas/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Precursores del ARN/química , Precursores del ARN/genética , Precursores del ARN/metabolismo , Ribonucleasa III/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/metabolismo
12.
Nature ; 497(7448): 217-23, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23636326

RESUMEN

The mammalian target of rapamycin (mTOR), a phosphoinositide 3-kinase-related protein kinase, controls cell growth in response to nutrients and growth factors and is frequently deregulated in cancer. Here we report co-crystal structures of a complex of truncated mTOR and mammalian lethal with SEC13 protein 8 (mLST8) with an ATP transition state mimic and with ATP-site inhibitors. The structures reveal an intrinsically active kinase conformation, with catalytic residues and a catalytic mechanism remarkably similar to canonical protein kinases. The active site is highly recessed owing to the FKBP12-rapamycin-binding (FRB) domain and an inhibitory helix protruding from the catalytic cleft. mTOR-activating mutations map to the structural framework that holds these elements in place, indicating that the kinase is controlled by restricted access. In vitro biochemistry shows that the FRB domain acts as a gatekeeper, with its rapamycin-binding site interacting with substrates to grant them access to the restricted active site. Rapamycin-FKBP12 inhibits the kinase by directly blocking substrate recruitment and by further restricting active-site access. The structures also reveal active-site residues and conformational changes that underlie inhibitor potency and specificity.


Asunto(s)
Serina-Treonina Quinasas TOR/química , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Furanos/química , Furanos/farmacología , Humanos , Indoles/química , Indoles/metabolismo , Indoles/farmacología , Magnesio/química , Magnesio/metabolismo , Modelos Moleculares , Naftiridinas/química , Naftiridinas/metabolismo , Naftiridinas/farmacología , Estructura Terciaria de Proteína/efectos de los fármacos , Purinas/química , Purinas/metabolismo , Purinas/farmacología , Piridinas/química , Piridinas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/química , Sirolimus/metabolismo , Sirolimus/farmacología , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteína 1A de Unión a Tacrolimus/química , Proteína 1A de Unión a Tacrolimus/metabolismo , Proteína 1A de Unión a Tacrolimus/farmacología , Homóloga LST8 de la Proteína Asociada al mTOR
13.
Proteins ; 86(3): 344-353, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29243286

RESUMEN

Protein kinase, casein kinase II (CK2), is ubiquitously expressed and highly conserved protein kinase that shows constitutive activity. It phosphorylates a diverse set of proteins and plays crucial role in several cellular processes. The catalytic subunit of this enzyme (CK2α) shows remarkable flexibility as evidenced in numerous crystal structures determined till now. Here, using analysis of multiple crystal structures and long timescale molecular dynamics simulations, we explore the conformational flexibility of CK2α. The enzyme shows considerably higher flexibility in the solution as compared to that observed in crystal structure ensemble. Multiple conformations of hinge region, located near the active site, were observed during the dynamics. We further observed that among these multiple conformations, the most populated conformational state was inadequately represented in the crystal structure ensemble. The catalytic spine, was found to be less dismantled in this state as compared to the "open" hinge/αD state crystal structures. The comparison of dynamics in unbound (Apo) state and inhibitor (CX4945) bound state exhibits inhibitor induced suppression in the overall dynamics of the enzyme. This is especially true for functionally important glycine-rich loop above the active site. Together, this work gives novel insights into the dynamics of CK2α in solution and relates it to the function. This work also explains the effect of inhibitor on the dynamics of CK2α and paves way for development of better inhibitors.


Asunto(s)
Dominio Catalítico , Simulación de Dinámica Molecular , Conformación Proteica , Aminoácidos/química , Aminoácidos/metabolismo , Sitios de Unión , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/química , Quinasa de la Caseína II/metabolismo , Cristalografía por Rayos X , Humanos , Naftiridinas/química , Naftiridinas/metabolismo , Naftiridinas/farmacología , Fenazinas , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
14.
Drug Metab Dispos ; 46(11): 1546-1555, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30171161

RESUMEN

Mass balance and biotransformation of finerenone, a nonsteroidal mineralocorticoid receptor antagonist, were investigated in four healthy male volunteers following a single oral administration of 10 mg (78 µCi) of [14C]finerenone and compared with data from studies in dogs and rats. The total recovery of the administered radioactivity was 101% in humans, 94.7% in dogs, and 95.2% in rats. In humans, radioactivity was mainly excreted renally (80%); in rats, it was primarily the biliary/fecal route (76%); and in dogs, excretion was more balanced. Finerenone was extensively metabolized in all species by oxidative biotransformation, with minor amounts of unchanged drug in excreta (humans: 1%; dogs, rats: <9%). In vitro studies suggested cytochrome P450 3A4 was the predominant enzyme involved in finerenone metabolism in humans. Primary metabolic transformation involved aromatization of the dihydronaphthyridine moiety of metabolite M1 as a major clearance pathway with a second oxidative pathway leading to M4. These were both prone to further oxidative biotransformation reactions. Naphthyridine metabolites (M1-M3) were the dominant metabolites identified in human plasma, with no on-target pharmacological activity. In dog plasma, finerenone and metabolite M2 constituted the major components; finerenone accounted almost exclusively for drug-related material in rat plasma. For metabolites M1-M3, axial chirality was observed, represented by two atropisomers (e.g., M1a and M1b). Analysis of plasma and excreta showed one atropisomer (a-series, >79%) of each metabolite predominated in all three species. In summary, the present study demonstrates that finerenone is cleared by oxidative biotransformation, mainly via naphthyridine derivatives.


Asunto(s)
Biotransformación/fisiología , Antagonistas de Receptores de Mineralocorticoides/metabolismo , Naftiridinas/metabolismo , Administración Oral , Anciano , Animales , Bilis/metabolismo , Citocromo P-450 CYP3A/metabolismo , Perros , Heces/química , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Ratas , Ratas Wistar
15.
Mol Pharmacol ; 92(3): 297-309, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28663279

RESUMEN

The α-like octopamine receptors (OctR) are believed to be the evolutionary precursor to the vertebrate α2-adrenergic receptors (α2-ARs) based upon sequence similarity and the ability to interact with norepinephrine and a number of compounds that bind with high affinity to α2-ARs. Barnacles and fruit flies are two prominent model marine and terrestrial representatives of the Arthropoda phylum, and although α-like OctRs have been cloned from Balanus improvisus (BiOctR) and Drosophila melanogaster (DmOctR), little is known about the structure-activity space for these important species. A diverse panel of 22 probes spanning different structural classes were employed to interrogate the structure-activity of the BiOctR and DmOctR. While BiOctR and DmOctR exhibited similar functional profiles for mammalian biogenic amine G protein-coupled receptor agonists and antagonists, some ligands had dramatically different mechanisms of action. For instance, significant differences in the efficacy for some agonists were observed, including that vertebrate biogenic amines structurally related to octopamine acted as superagonists at the DmOctR but partial agonists at the BiOctR, and the two species diverged in their sensitivities to the α2-AR antagonist [3H]rauwolscine. Furthermore, sodium enhanced [3H]rauwolscine's interactions with the BiOctR, but not at a vertebrate α2-AR. Molecular mechanistic studies indicate that rauwolscine interacts with the BiOctR, DmOctR, and α2C-adrenergic receptor at an allosteric site. In addition, compounds that acted as agonists at a cloned α-like BiOctR also induced a hyperactivity response in Balanus cyprids mediated by the α-like OctR, suggesting that the receptor may serve as a higher throughput proxy for discovering compounds with potential cyprid deterrent properties.


Asunto(s)
Receptores de Amina Biogénica/química , Receptores de Amina Biogénica/fisiología , Thoracica/química , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Drosophila melanogaster , Células HEK293 , Humanos , Isoquinolinas/metabolismo , Naftiridinas/metabolismo , Filogenia , Receptores de Amina Biogénica/agonistas , Sodio/farmacología , Relación Estructura-Actividad , Thoracica/genética
16.
Proc Natl Acad Sci U S A ; 111(49): 17450-5, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25422441

RESUMEN

Anabolic and catabolic signaling oppose one another in adipose tissue to maintain cellular and organismal homeostasis, but these pathways are often dysregulated in metabolic disorders. Although it has long been established that stimulation of the ß-adrenergic receptor inhibits insulin-stimulated glucose uptake in adipocytes, the mechanism has remained unclear. Here we report that ß-adrenergic-mediated inhibition of glucose uptake requires lipolysis. We also show that lipolysis suppresses glucose uptake by inhibiting the mammalian target of rapamycin (mTOR) complexes 1 and 2 through complex dissociation. In addition, we show that products of lipolysis inhibit mTOR through complex dissociation in vitro. These findings reveal a previously unrecognized intracellular signaling mechanism whereby lipolysis blocks the phosphoinositide 3-kinase-Akt-mTOR pathway, resulting in decreased glucose uptake. This previously unidentified mechanism of mTOR regulation likely contributes to the development of insulin resistance.


Asunto(s)
Adipocitos/citología , Catecolaminas/química , Glucosa/farmacocinética , Lipólisis/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Células 3T3-L1 , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Homeostasis , Hiperglucemia/metabolismo , Insulina/metabolismo , Resistencia a la Insulina , Lípidos/química , Ratones , Modelos Biológicos , Naftiridinas/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal
17.
Genes Cells ; 20(4): 292-309, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25651869

RESUMEN

Rapamycin and its derivatives have now emerged as an attractive therapeutic strategy with both immunosuppressant and antitumor properties. In addition, rapamycin has been proposed as a calorie restriction mimetic to extend the life span of various organisms. The fission yeast Schizosaccharomyces pombe (S. pombe) serves as a valuable genetic model system to study the mechanism(s) of drug action as well as to determine genetic contexts associated with drug sensitivity or resistance. Here, we identified genes that when deleted modulate the rapamycin-sensitive strains in S. pombe. We carried out a chemical genomics screen for rapamycin-sensitive mutants using the genome-deletion library which covers 95.3% of all nonessential fission yeast genes and confirmed 59 genes to be rapamycin sensitive. Gene Ontology (GO) enrichment analysis showed that strains sensitive to rapamycin are highly enriched in processes regulating tRNA modification and mitochondria as well as other ontologies, including cellular metabolic process, chromatin organization, cell cycle, signaling, translation, transport and other cellular processes. Analysis also showed that components of the Elongator complex are overrepresented in the sensitive strains. Here, the data obtained will provide valuable information for speculation on the actions of rapamycin as well as on TORC signaling, thereby presenting a strategy to enhance sensitivity to rapamycin.


Asunto(s)
Antifúngicos/metabolismo , Farmacorresistencia Fúngica , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Sirolimus/metabolismo , Ciclo Celular , Cromatina/genética , Genoma Fúngico , Genómica/métodos , Mitocondrias/genética , Mutación , Naftiridinas/metabolismo , Biosíntesis de Proteínas , Inhibidores de Proteínas Quinasas/metabolismo , ARN de Transferencia/genética , Schizosaccharomyces/citología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
18.
Xenobiotica ; 46(9): 784-92, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26711252

RESUMEN

1. Interspecies allometry scaling for prediction of human excretory amounts in urine or feces was performed for numerous antibacterials. Antibacterials used for urinary scaling were: rifapentine, pefloxacin, trovafloxacin (Gr1/low; <10%); miloxacin, linezolid, PNU-142300 (Gr2/medium; 10-40%); aztreonam, carumonam, cefozopran, doripenem, imipenem, and ceftazidime (Gr3/high; >50%). Rifapentine, cabotegravir, and dolutegravir was used for fecal scaling (high; >50%). 2. The employment of allometry equation: Y = aW(b) enabled scaling of urine/fecal amounts from animal species. Corresponding predicted amounts were converted into % recovery by considering the respective human dose. Comparison of predicted/observed values enabled fold difference and error calculations (mean absolute error [MAE] and root mean square error [RMSE]). Comparisons were made for urinary/fecal data; and qualitative assessment was made amongst Gr1/Gr2/Gr3 for urine. 3. Average correlation coefficient for the allometry scaling was >0.995. Excretory amount predictions were largely within 0.75- to 1.5-fold differences. Average MAE and RMSE were within ±22% and 23%, respectively. Although robust predictions were achieved for higher urinary/fecal excretion (>50%), interspecies scaling was applicable for low/medium excretory drugs. 4. Based on the data, interspecies scaling of urine or fecal excretory amounts may be potentially used as a tool to understand the significance of either urinary or fecal routes of elimination in humans in early development.


Asunto(s)
Antibacterianos/metabolismo , Animales , Antibacterianos/orina , Aztreonam/análogos & derivados , Aztreonam/metabolismo , Carbapenémicos/metabolismo , Ceftazidima/metabolismo , Cefalosporinas/metabolismo , Doripenem , Heces/química , Fluoroquinolonas/metabolismo , Compuestos Heterocíclicos con 3 Anillos/metabolismo , Humanos , Imipenem/metabolismo , Linezolid/metabolismo , Naftiridinas/metabolismo , Oxazinas , Ácido Oxolínico/análogos & derivados , Ácido Oxolínico/metabolismo , Pefloxacina/metabolismo , Piperazinas , Piridonas , Estudios Retrospectivos , Rifampin/análogos & derivados , Rifampin/metabolismo , Cefozoprán
19.
Drug Metab Dispos ; 43(7): 928-35, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25870101

RESUMEN

OTS167 is a potent maternal embryonic leucine zipper kinase inhibitor undergoing clinical testing as antineoplastic agent. We aimed to identify the UDP-glucuronosyltransferases (UGTs) involved in OTS167 metabolism, study the relationship between UGT genetic polymorphisms and hepatic OTS167 glucuronidation, and investigate the inhibitory potential of OTS167 on UGTs. Formation of a single OTS167-glucuronide (OTS167-G) was observed in pooled human liver (HLM) (Km = 3.4 ± 0.2 µM), intestinal microsomes (HIM) (Km = 1.7 ± 0.1 µM), and UGTs. UGT1A1 (64 µl/min/mg) and UGT1A8 (72 µl/min/mg) exhibited the highest intrinsic clearances (CLint) for OTS167, followed by UGT1A3 (51 µl/min/mg) and UGT1A10 (47 µl/min/mg); UGT1A9 was a minor contributor. OTS167 glucuronidation in HLM was highly correlated with thyroxine glucuronidation (r = 0.91, P < 0.0001), SN-38 glucuronidation (r = 0.79, P < 0.0001), and UGT1A1 mRNA (r = 0.72, P < 0.0001). Nilotinib (UGT1A1 inhibitor) and emodin (UGT1A8 and UGT1A10 inhibitor) exhibited the highest inhibitory effects on OTS167-G formation in HLM (68%) and HIM (47%). We hypothesize that OTS167-G is an N-glucuronide according to mass spectrometry. A significant association was found between rs6706232 and reduced OTS167-G formation (P = 0.03). No or weak UGT inhibition (range: 0-21%) was observed using clinically relevant OTS167 concentrations (0.4-2 µM). We conclude that UGT1A1 and UGT1A3 are the main UGTs responsible for hepatic formation of OTS167-G. Intestinal UGT1A1, UGT1A8, and UGT1A10 may contribute to first-pass OTS167 metabolism after oral administration.


Asunto(s)
Antineoplásicos/metabolismo , Glucuronosiltransferasa/metabolismo , Naftiridinas/metabolismo , Inhibidores Enzimáticos/farmacología , Genotipo , Glucurónidos/metabolismo , Glucuronosiltransferasa/antagonistas & inhibidores , Glucuronosiltransferasa/genética , Humanos , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Hígado/enzimología , Microsomas/enzimología , Microsomas Hepáticos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Tiroxina/metabolismo
20.
Chemistry ; 21(30): 10688-95, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26177045

RESUMEN

In vitro replication of an unnatural imidazopyridopyridine:naphthyridine base pair, (i.e., ImN(N):NaO(O)), having four hydrogen bonds was investigated. Kinetic studies of single-nucleotide insertion revealed that ImN(N) and NaO(O) were recognized as complementary bases by an exonuclease-deficient Klenow fragment with higher specificity and efficiency than two previously described pairs (ImN(O):NaO(N) and ImO(N):NaN(O)) because of higher thermal and thermodynamic stabilities and the DAAD:ADDA (D=donor, A=acceptor) hydrogen-bonding pattern of the ImN(N):NaO(O) pair. Faithful polymerase chain reaction (PCR) amplification of a DNA fragment containing the ImN(N):NaO(O) pair was achieved by using DNA polymerases possessing 3'→5' exonuclease activity (≈99.5 % per doubling).


Asunto(s)
Emparejamiento Base , ADN/química , Naftiridinas/química , Reacción en Cadena de la Polimerasa , Piridinas/química , Secuencia de Bases , ADN/genética , ADN/metabolismo , ADN Polimerasa I/metabolismo , Enlace de Hidrógeno , Imidazoles/química , Imidazoles/metabolismo , Cinética , Modelos Moleculares , Naftiridinas/metabolismo , Piridinas/metabolismo , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA