Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(3): 636-648.e13, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30682372

RESUMEN

Despite intensive efforts to discover highly effective treatments to eradicate tuberculosis (TB), it remains as a major threat to global human health. For this reason, new TB drugs directed toward new targets are highly coveted. MmpLs (Mycobacterial membrane proteins Large), which play crucial roles in transporting lipids, polymers and immunomodulators and which also extrude therapeutic drugs, are among the most important therapeutic drug targets to emerge in recent times. Here, crystal structures of mycobacterial MmpL3 alone and in complex with four TB drug candidates, including SQ109 (in Phase 2b-3 clinical trials), are reported. MmpL3 consists of a periplasmic pore domain and a twelve-helix transmembrane domain. Two Asp-Tyr pairs centrally located in this domain appear to be key facilitators of proton-translocation. SQ109, AU1235, ICA38, and rimonabant bind inside the transmembrane region and disrupt these Asp-Tyr pairs. This structural data will greatly advance the development of MmpL3 inhibitors as new TB drugs.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/ultraestructura , Adamantano/análogos & derivados , Adamantano/metabolismo , Antituberculosos/química , Transporte Biológico , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Etilenodiaminas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/ultraestructura , Compuestos de Fenilurea/metabolismo , Rimonabant/metabolismo , Tuberculosis/microbiología
2.
Molecules ; 27(20)2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36296721

RESUMEN

Tuberculosis remains an important cause of morbidity and mortality throughout the world. Notably, an important number of multi drug resistant cases is an increasing concern. This problem points to an urgent need for novel compounds with antimycobacterial properties and to improve existing therapies. Whole-cell-based screening for compounds with activity against Mycobacterium tuberculosis complex strains in the presence of linezolid was performed in this study. A set of 15 bioactive compounds with antimycobacterial activity in vitro were identified with a minimal inhibitory concentration of less than 2 µg/mL. Among them, compound 1 is a small molecule with a chemical structure consisting of an adamantane moiety and a hydrazide-hydrazone moiety. Whole genome sequencing of spontaneous mutants resistant to the compounds suggested compound 1 to be a new inhibitor of MmpL3. This compound binds to the same pocket as other already published MmpL3 inhibitors, without disturbing the proton motive force of M. bovis BCG and M. smegmatis. Compound 1 showed a strong activity against a panel ofclinical strains of M. tuberculosis in vitro. This compound showed no toxicity against mammalian cells and protected Galleria mellonella larvae against M. bovis BCG infection. These results suggest that compound 1 is a promising anti-TB agent with the potential to improve TB treatment in combination with standard TB therapies.


Asunto(s)
Adamantano , Mycobacterium tuberculosis , Tuberculosis , Animales , Humanos , Antituberculosos/uso terapéutico , Hidrazonas/farmacología , Hidrazonas/uso terapéutico , Linezolid/metabolismo , Vacuna BCG/metabolismo , Vacuna BCG/uso terapéutico , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/genética , Pruebas de Sensibilidad Microbiana , Tuberculosis/tratamiento farmacológico , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Adamantano/farmacología , Adamantano/metabolismo , Mamíferos/metabolismo
3.
J Gen Virol ; 102(9)2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34546870

RESUMEN

Tick-borne encephalitis virus (TBEV), a member of the genus Flavivirus, is common in Europe and Asia and causes a severe disease of the central nervous system. A promising approach in the development of therapy for TBEV infection is the search for small molecule antivirals targeting the flavivirus envelope protein E, particularly its ß-n-octyl-d-glucoside binding pocket (ß-OG pocket). However, experimental studies of candidate antivirals may be complicated by varying amounts and different forms of the protein E in the virus samples. Viral particles with different conformations and arrangements of the protein E are produced during the replication cycle of flaviviruses, including mature, partially mature, and immature forms, as well as subviral particles lacking genomic RNA. The immature forms are known to be abundant in the viral population. We obtained immature virion preparations of TBEV, characterized them by RT-qPCR, and assessed in vivo and in vitro infectivity of the residual mature virions in the immature virus samples. Analysis of the ß-OG pocket structure on the immature virions confirmed the possibility of binding of adamantylmethyl esters of 5-aminoisoxazole-3-carboxylic acid in the pocket. We demonstrated that the antiviral activity of these compounds in plaque reduction assay is significantly reduced in the presence of immature TBEV particles.


Asunto(s)
Adamantano/farmacología , Antivirales/farmacología , Virus de la Encefalitis Transmitidos por Garrapatas/efectos de los fármacos , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Encefalitis Transmitida por Garrapatas/virología , Isoxazoles/farmacología , Virión/fisiología , Adamantano/metabolismo , Animales , Antivirales/metabolismo , Línea Celular , Virus de la Encefalitis Transmitidos por Garrapatas/crecimiento & desarrollo , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Glucósidos/metabolismo , Isoxazoles/metabolismo , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Porcinos , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Ensayo de Placa Viral , Virión/inmunología , Virión/patogenicidad , Virión/ultraestructura
4.
Inorg Chem ; 60(23): 17783-17796, 2021 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-34762413

RESUMEN

PtmU3 is a newly identified nonheme diiron monooxygenase, which installs a C-5 ß-hydroxyl group into the C-19 CoA-ester intermediate involved in the biosynthesis of unique diterpene-derived scaffolds of platensimycin and platencin. PtmU3 possesses a noncanonical diiron active site architecture of a saturated six-coordinate iron center and lacks the µ-oxo bridge. Although the hydroxylation process is a simple reaction for nonheme mononuclear iron-dependent enzymes, how PtmU3 employs the diiron center to catalyze the H-abstraction and OH-rebound is still unknown. In particular, the electronic characteristic of diiron is also unclear. To understand the catalytic mechanism of PtmU3, we constructed two reactant models in which both the Fe1II-Fe2III-superoxo and Fe1II-Fe2IV═O are considered to trigger the H-abstraction and performed a series of quantum mechanics/molecular mechanics calculations. Our calculation results reveal that PtmU3 is a special monooxygenase, that is, both atoms of the dioxygen molecule can be incorporated into two molecules of the substrate by the successive reactions. In the first-round reaction, PtmU3 uses the Fe1II-Fe2III-superoxo to install a hydroxyl group into the substrate, generating the high-reactive Fe1II-Fe2IV═O complex. In the second-round reaction, the Fe1II-Fe2IV═O species is responsible for the hydroxylation of another molecule of the substrate. In the diiron center, Fe2 adopts the high spin state (S = 5/2) during the catalysis, whereas for Fe1, in addition to its structural role, it may also play an assistant role for Fe1 catalysis. In the two successive OH-installing steps, the H-abstraction is always the rate-liming step. E241 and D308 not only act as bridging ligands to connect two Fe ions but also take part in the electron reorganization. Owing to the high reactivity of Fe1II-Fe2IV═O compared to Fe1II-Fe2III-superoxo, besides the C5-hydroxylation, the C3- or C18-hydroxylation was also calculated to be feasible.


Asunto(s)
Adamantano/metabolismo , Aminobenzoatos/metabolismo , Anilidas/metabolismo , Teoría Funcional de la Densidad , Oxigenasas de Función Mixta/metabolismo , Simulación de Dinámica Molecular , Adamantano/química , Aminobenzoatos/química , Anilidas/química , Biocatálisis , Hidroxilación , Estructura Molecular
5.
Molecules ; 26(8)2021 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-33921487

RESUMEN

LW6, an (aryloxyacetylamino)benzoic acid derivative, was recently identified to be an inhibitor of hypoxia-inducible factor-1α (HIF-1α), which is an attractive target for cancer therapeutics. Although LW6 is known to act by inhibiting the accumulation of HIF-1α, pharmacokinetics needs to be evaluated to assess its potential as an anti-tumor agent. Here, we investigated the plasma pharmacokinetics and metabolism of LW6 in mice. LW6 exhibited a small volume of distribution (0.5 ± 0.1 L/kg), and a short terminal half-life (0.6 ± 0.1 h). Following intravenous or oral administration, LW6 was rapidly converted to its active metabolite, (4-adamantan-1-yl-phenoxy)acetic acid (APA). Although LW6 was rapidly absorbed, its oral bioavailability, estimated using AUClast values, was low (1.7 ± 1.8%). It was slowly degraded in mouse liver microsomes (t1/2 > 1 h) and serum (t1/2 > 6 h). About 54% or 44.8% of LW6 was available systemically as APA in the mouse after a single intravenous or oral administration, respectively. Thus, our results indicated the need to simultaneously consider the active metabolite as well as the parent compound for successful evaluation during lead optimization.


Asunto(s)
Acetanilidas/farmacología , Acetanilidas/farmacocinética , Adamantano/análogos & derivados , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Acetanilidas/sangre , Acetanilidas/metabolismo , Adamantano/sangre , Adamantano/metabolismo , Adamantano/farmacocinética , Adamantano/farmacología , Animales , Células CACO-2 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inyecciones Intravenosas , Masculino , Metaboloma , Ratones Endogámicos ICR , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Factores de Tiempo
6.
Molecules ; 26(7)2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33808438

RESUMEN

A novel HIF (hypoxia-inducible factor)-1α inhibitor, the (aryloxyacetylamino)benzoic acid derivative LW6, is an anticancer agent that inhibits the accumulation of HIF-1α. The aim of this study was to characterize and determine the structures of the metabolites of LW6 in ICR mice. Metabolite identification was performed using a predictive multiple reaction monitoring-information dependent acquisition-enhanced product ion (pMRM-IDA-EPI) method in negative ion mode on a hybrid triple quadrupole-linear ion trap mass spectrometer (QTRAP). A total of 12 metabolites were characterized based on their MS/MS spectra, and the retention times were compared with those of the parent compound. The metabolites were divided into five structural classes based on biotransformation reactions: amide hydrolysis, ester hydrolysis, mono-oxidation, glucuronidation, and a combination of these reactions. From this study, 2-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)acetic acid (APA, M7), the metabolite produced via amide hydrolysis, was found to be a major circulating metabolite of LW6 in mice. The results of this study can be used to improve the pharmacokinetic profile by lowering the clearance and increasing the exposure relative to LW6.


Asunto(s)
Acetanilidas , Adamantano/análogos & derivados , Antineoplásicos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Acetanilidas/sangre , Acetanilidas/metabolismo , Acetanilidas/farmacocinética , Adamantano/sangre , Adamantano/metabolismo , Adamantano/farmacocinética , Animales , Antineoplásicos/sangre , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Biotransformación , Masculino , Ratones , Ratones Endogámicos ICR
7.
Angew Chem Int Ed Engl ; 60(10): 5429-5435, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33247547

RESUMEN

Hierarchical self-assembly (HAS) is a powerful approach to create supramolecular nanostructures for biomedical applications. This potency, however, is generally challenged by the difficulty of controlling the HAS of biomacromolecules and the functionality of resulted HAS nanostructures. Herein, we report a modular approach for controlling the HAS of discrete metal-organic cages (MOC) into supramolecular nanoparticles, and its potential for intracellular protein delivery and cell-fate specification. The hierarchical coordination-driven self-assembly of adamantane-functionalized M12 L24 MOC (Ada-MOC) and the host-guest interaction of Ada-MOC with ß-cyclodextrin-conjugated polyethylenimine (PEI-ßCD) afford supramolecular nanoparticles in a controllable manner. HAS maintains high efficiency and orthogonality in the presence of protein, enabling the encapsulation of protein into the nanoparticles for intracellular protein delivery for therapeutic application and CRISPR/Cas9 genome editing.


Asunto(s)
Portadores de Fármacos/química , Estructuras Metalorgánicas/química , Nanopartículas/química , Adamantano/análogos & derivados , Adamantano/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Portadores de Fármacos/síntesis química , Portadores de Fármacos/metabolismo , Endocitosis/fisiología , Edición Génica/métodos , Genoma Humano , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Integrasas/genética , Integrasas/metabolismo , Estructuras Metalorgánicas/síntesis química , Estructuras Metalorgánicas/metabolismo , Nanopartículas/metabolismo , Polietileneimina/análogos & derivados , Polietileneimina/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Ribonucleasa Pancreática/metabolismo , Ribonucleasa Pancreática/farmacología , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , beta-Ciclodextrinas/síntesis química , beta-Ciclodextrinas/química , beta-Ciclodextrinas/metabolismo
8.
J Biol Chem ; 294(45): 16567-16576, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31570525

RESUMEN

Throughout history, natural products have significantly contributed to the discovery of novel chemistry, drug leads, and tool molecules to probe and address complex challenges in biology and medicine. Recent microbial genome sequencing efforts have uncovered many microbial biosynthetic gene clusters without an associated natural product. This means that the natural products isolated to date do not fully reflect the biosynthetic potential of microbial strains. This observation has rejuvenated the natural product community and inspired a return to microbial strain collections. Mining large microbial strain collections with the most current technologies in genome sequencing, bioinformatics, and high-throughput screening techniques presents new opportunities in natural product discovery. In this review, we report on the newly expanded microbial strain collection at The Scripps Research Institute, which represents one of the largest and most diverse strain collections in the world. Two complementary approaches, i.e. structure-centric and function-centric, are presented here to showcase how to leverage a large microbial strain collection for natural product discovery and to address challenges and harness opportunities for future efforts. Highlighted examples include the discovery of alternative producers of known natural products with superior growth characteristics and high titers, novel analogs of privileged scaffolds, novel natural products, and new activities of known and new natural products. We anticipate that this large microbial strain collection will facilitate the discovery of new natural products for many applications.


Asunto(s)
Productos Biológicos/metabolismo , Adamantano/química , Adamantano/metabolismo , Aminobenzoatos/química , Aminobenzoatos/metabolismo , Anilidas/química , Anilidas/metabolismo , Bacterias/genética , Bacterias/metabolismo , Productos Biológicos/química , Biología Computacional/métodos , Bases de Datos Genéticas , Hongos/genética , Hongos/metabolismo , Genoma Bacteriano , Familia de Multigenes
9.
Bioorg Med Chem Lett ; 30(18): 127430, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32736212

RESUMEN

A series of inhibitors of the soluble epoxide hydrolase (sEH) containing lipophilic groups of natural origin (camphanyl, norcamphanyl, furan-2-yl) were developed. Inhibitory potency ranging from 0.4 nM to 2.16 µM were obtained. While having the same level of inhibitory activity bicyclic ureas are up to 10-fold more soluble than the corresponding ureas containing adamantyl or 4-trifluoromethoxyphenyl substituents. This makes them easier to formulate, more bioavailable and thus more promising as therapeutic sEH inhibitors. Endo/exo-form of compound 2b derived from l-camphor is 14-fold more potent than the corresponding analogue derived from d-camphor (IC50 = 3.7 nM vs. 50.6 nM) indicating enantiomeric preference.


Asunto(s)
Adamantano/química , Inhibidores Enzimáticos/química , Epóxido Hidrolasas/antagonistas & inhibidores , Lípidos/química , Adamantano/metabolismo , Sitios de Unión , Inhibidores Enzimáticos/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad , Urea/análogos & derivados , Urea/química , Agua
10.
Bioorg Med Chem Lett ; 30(3): 126908, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31870649

RESUMEN

A series of inhibitors of the soluble epoxide hydrolase (sEH) containing imidazolidine-2,4,5-trione or pirimidine-2,4,6-trione has been synthesized. Inhibition potency of the described compounds ranges from 8.4 µM to 0.4 nM. The tested compounds possess higher water solubility than their preceding ureas. Molecular docking indicates new bond between the triones and the active site of sEH that in part explain the observed potency of the new pharmacophores. While less potent than the corresponding ureas, the modifications of urea group reported herein yield compounds with higher water solubility, thus permitting easier formulation.


Asunto(s)
Inhibidores Enzimáticos/química , Epóxido Hidrolasas/antagonistas & inhibidores , Imidazolidinas/química , Pirimidinas/química , Adamantano/química , Adamantano/metabolismo , Sitios de Unión , Dominio Catalítico , Inhibidores Enzimáticos/metabolismo , Epóxido Hidrolasas/metabolismo , Humanos , Imidazolidinas/metabolismo , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Pirimidinas/metabolismo , Solubilidad , Relación Estructura-Actividad , Urea/química , Urea/metabolismo
11.
Xenobiotica ; 50(7): 863-874, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31791185

RESUMEN

1. Treatment periods of P-glycoprotein (P-gp) inhibitors have revealed different efficacies. We have previously reported dose-dependent inhibition of P-gp in single-treatment with LC478. However, whether repeated treatment with LC478 can inhibit P-gp even at its ineffective single-treatment dose remains unknown. 2. Therefore, the purpose of this study was to assess the effect of repeated treatment (i.e., 7-day treatment) with LC478 on P-gp known to affect docetaxel bioavailability in rats. Effects of LC478 on P-gp mediated efflux and expression in MDCK-MDR1 cells, P-gp ATPase activity, and binding site with P-gp were evaluated.3. The 7-day treatment with LC478 increased docetaxel absorption via intestinal P-gp inhibition in rats. Intestinal concentrations of LC478 were 8.31-10.3 µM in rats after 7-day treatment of LC478. These concentrations were close to 10 µM that reduced P-gp mediated docetaxel efflux and P-gp expression in MDCK-MDR1 cells. Considering that intestinal LC478 concentrations after 1-day treatment were 2.68-4.19 µM, higher LC478 concentrations after 7-day treatment might have driven P-gp inhibition and increased docetaxel absorption. LC478 might competitively inhibit P-gp considering its stimulated ATPase activity and its binding site with nucleotide binding domain of P-gp. 4. Therefore, repeated treatment with LC478 can determine its feasibility for P-gp inhibition and changing docetaxel bioavailability.


Asunto(s)
Adamantano/análogos & derivados , Adamantano/metabolismo , Antineoplásicos/farmacocinética , Docetaxel/farmacocinética , Inhibidores Enzimáticos/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adamantano/farmacocinética , Animales , Disponibilidad Biológica , Transporte Biológico , Absorción Intestinal , Ratas
12.
J Am Chem Soc ; 141(31): 12406-12412, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31291107

RESUMEN

Nonheme diiron monooxygenases make up a rapidly growing family of oxygenases that are rarely identified in secondary metabolism. Herein, we report the in vivo, in vitro, and structural characterizations of a nonheme diiron monooxygenase, PtmU3, that installs a C-5 ß-hydroxyl group in the unified biosynthesis of platensimycin and platencin, two highly functionalized diterpenoids that act as potent and selective inhibitors of bacterial and mammalian fatty acid synthases. This hydroxylation sets the stage for the subsequent A-ring cleavage step key to the unique diterpene-derived scaffolds of platensimycin and platencin. PtmU3 adopts an unprecedented triosephosphate isomerase (TIM) barrel structural fold for this class of enzymes and possesses a noncanonical diiron active site architecture with a saturated six-coordinate iron center lacking a µ-oxo bridge. This study reveals the first member of a previously unidentified superfamily of TIM-barrel-fold enzymes for metal-dependent dioxygen activation, with the majority predicted to act on CoA-linked substrates, thus expanding our knowledge of nature's repertoire of nonheme diiron monooxygenases and TIM-barrel-fold enzymes.


Asunto(s)
Adamantano/metabolismo , Aminobenzoatos/metabolismo , Aminofenoles/metabolismo , Anilidas/metabolismo , Hierro/metabolismo , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/metabolismo , Compuestos Policíclicos/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Hidroxilación , Modelos Moleculares
13.
J Am Chem Soc ; 141(9): 4043-4050, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30735041

RESUMEN

Platensimycin (PTM) and platencin (PTN) are highly functionalized bacterial diterpenoids of ent-kauranol and ent-atiserene biosynthetic origin. C7 oxidation in the B-ring plays a key biosynthetic role in generating structural complexity known for ent-kaurane and ent-atisane derived diterpenoids. While all three oxidation patterns, α-hydroxyl, ß-hydroxyl, and ketone, at C7 are seen in both the ent-kaurane and ent-atisane derived diterpenoids, their biosynthetic origins remain largely unknown. We previously established that PTM and PTN are produced by a single biosynthetic machinery, featuring cryptic C7 oxidations at the B-rings that transform the ent-kauranol and ent-atiserene derived precursors into the characteristic PTM and PTN scaffolds. Here, we report a three-enzyme cascade affording C7 α-hydroxylation in PTM and PTN biosynthesis. Combining in vitro and in vivo studies, we show that PtmO3 and PtmO6 are two functionally redundant α-ketoglutarate-dependent dioxygenases that generate a cryptic C7 ß-hydroxyl on each of the ent-kauranol and ent-atiserene scaffolds, and PtmO8 and PtmO1, a pair of NAD+/NADPH-dependent dehydrogenases, subsequently work in concert to invert the C7 ß-hydroxyl to α-hydroxyl via a C7 ketone intermediate. PtmO3 and PtmO6 represent the first dedicated C7 ß-hydroxylases characterized to date and, together with PtmO8 and PtmO1, provide an account for the biosynthetic origins of all three C7 oxidation patterns that may shed light on other B-ring modifications in bacterial, plant, and fungal diterpenoid biosynthesis. Given their unprecedented activities in C7 oxidations, PtmO3, PtmO6, PtmO8, and PtmO1 enrich the growing toolbox of novel enzymes that could be exploited as biocatalysts to rapidly access complex diterpenoid natural products.


Asunto(s)
Adamantano/metabolismo , Aminobenzoatos/metabolismo , Aminofenoles/metabolismo , Anilidas/metabolismo , Compuestos Policíclicos/metabolismo , Adamantano/química , Aminobenzoatos/química , Aminofenoles/química , Anilidas/química , Hidroxilación , Conformación Molecular , Oxidación-Reducción , Compuestos Policíclicos/química , Estereoisomerismo
14.
J Am Chem Soc ; 141(29): 11481-11488, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31184871

RESUMEN

The amantadine-resistant S31N mutant of the influenza A M2 proton channel has become prevalent in currently circulating viruses. Here, we have solved an X-ray crystal structure of M2(22-46) S31N that contains two distinct conformational states within its asymmetric unit. This structure reveals the mechanism of adamantane resistance in both conformational states of the M2 channel. In the Inwardopen conformation, the mutant Asn31 side chain faces the channel pore and sterically blocks the adamantane binding site. In the Inwardclosed conformation, Asn31 forms hydrogen bonds with carbonyls at the monomer-monomer interface, which twists the monomer helices and constricts the channel pore at the drug binding site. We also examine M2(19-49) WT and S31N using solution NMR spectroscopy and show that distribution of the two conformational states is dependent on both detergent choice and experimental pH.


Asunto(s)
Virus de la Influenza A/química , Virus de la Influenza A/genética , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética , Adamantano/metabolismo , Adamantano/farmacología , Amantadina/farmacología , Asparagina/química , Asparagina/genética , Asparagina/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Farmacorresistencia Viral/genética , Enlace de Hidrógeno , Concentración de Iones de Hidrógeno , Virus de la Influenza A/efectos de los fármacos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Proteínas de la Matriz Viral/metabolismo
15.
Nature ; 498(7455): 521-5, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23739335

RESUMEN

The hepatitis C virus (HCV) has developed a small membrane protein, p7, which remarkably can self-assemble into a large channel complex that selectively conducts cations. We wanted to examine the structural solution that the viroporin adopts in order to achieve selective cation conduction, because p7 has no homology with any of the known prokaryotic or eukaryotic channel proteins. The activity of p7 can be inhibited by amantadine and rimantadine, which are potent blockers of the influenza M2 channel and licensed drugs against influenza infections. The adamantane derivatives have been used in HCV clinical trials, but large variation in drug efficacy among the various HCV genotypes has been difficult to explain without detailed molecular structures. Here we determine the structures of this HCV viroporin as well as its drug-binding site using the latest nuclear magnetic resonance (NMR) technologies. The structure exhibits an unusual mode of hexameric assembly, where the individual p7 monomers, i, not only interact with their immediate neighbours, but also reach farther to associate with the i+2 and i+3 monomers, forming a sophisticated, funnel-like architecture. The structure also points to a mechanism of cation selection: an asparagine/histidine ring that constricts the narrow end of the funnel serves as a broad cation selectivity filter, whereas an arginine/lysine ring that defines the wide end of the funnel may selectively allow cation diffusion into the channel. Our functional investigation using whole-cell channel recording shows that these residues are critical for channel activity. NMR measurements of the channel-drug complex revealed six equivalent hydrophobic pockets between the peripheral and pore-forming helices to which amantadine or rimantadine binds, and compound binding specifically to this position may allosterically inhibit cation conduction by preventing the channel from opening. Our data provide a molecular explanation for p7-mediated cation conductance and its inhibition by adamantane derivatives.


Asunto(s)
Hepacivirus/química , Proteínas Virales/química , Adamantano/análogos & derivados , Adamantano/química , Adamantano/metabolismo , Adamantano/farmacología , Sitios de Unión , Difusión , Microscopía Electrónica , Modelos Biológicos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Porosidad , Rimantadina/química , Rimantadina/metabolismo , Rimantadina/farmacología , Relación Estructura-Actividad , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura
16.
J Ind Microbiol Biotechnol ; 46(3-4): 433-444, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30426283

RESUMEN

Recent advances and emerging technologies for metabolic pathway engineering and synthetic biology have transformed the field of natural product discovery, production, and engineering. Despite these advancements, there remain many challenges in understanding how biosynthetic gene clusters are silenced or activated, including changes in the transcription of key biosynthetic and regulatory genes. This knowledge gap is highlighted by the success and failed attempts of manipulating regulatory genes within biosynthetic gene clusters in both native producers and heterologous hosts. These complexities make the choice of native producers versus heterologous hosts, fermentation medium, and supply of precursors crucial factors in achieving the production of the target natural products and engineering designer analogs. Nature continues to serve as inspiration for filling the knowledge gaps and developing new research strategies. By exploiting the evolutionary power of nature, alternative producers, with the desired genetic amenability and higher titers of the target natural products, and new strains, harboring gene clusters that encode evolutionary optimized congeners of the targeted natural product scaffolds, can be discovered. These newly identified strains can serve as an outstanding biotechnology platform for the engineered production of sufficient quantities of the target natural products and their analogs, enabling biosynthetic studies and potential therapeutic applications. These challenges and opportunities are showcased herein using fredericamycin, iso-migrastatin, platencin and platensimycin, the enediynes of C-1027, tiancimycin, and yangpumicin, and the leinamycin family of natural products.


Asunto(s)
Productos Biológicos/química , Descubrimiento de Drogas , Adamantano/metabolismo , Aminobenzoatos/metabolismo , Aminoglicósidos/química , Aminofenoles/química , Anilidas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Vías Biosintéticas/genética , Enediinos/química , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Lactamas/química , Macrólidos/química , Ingeniería Metabólica , Familia de Multigenes , Piperidonas/química , Compuestos Policíclicos/química , Conformación Proteica , Análisis de Secuencia de ADN , Streptomyces/genética , Streptomyces/metabolismo , Tiazoles/química , Tionas/química
17.
J Am Chem Soc ; 140(39): 12349-12353, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30216060

RESUMEN

Platensimycin (PTM) and platencin (PTN) are potent and selective inhibitors of bacterial and mammalian fatty acid synthases. The regio- and stereospecificity of the ether oxygen atom in PTM, which PTN does not have, strongly contribute to the selectivity and potency of PTM. We previously reported the biosynthetic origin of the 11 S,16 S-ether moiety by characterizing the diterpene synthase PtmT3 as a (16 R)- ent-kauran-16-ol synthase and isolating 11-deoxy-16 R-hydroxylated congeners of PTM from the Δ ptmO5 mutant. PtmO5, a cytochrome P450, was proposed to catalyze formation of the ether moiety in PTM. Here we report the in vitro characterization of PtmO5, revealing that PtmO5 stereoselectively hydroxylates the C-11 position of the ent-kaurane scaffold resulting in an 11 S,16 R-diol intermediate. The ether moiety, the oxygen of which originates from the P450-catalyzed hydroxylation at C-11, is formed via cyclization of the diol intermediate. This study provides mechanistic insight into ether formation in natural product biosynthetic pathways.


Asunto(s)
Adamantano/metabolismo , Aminobenzoatos/metabolismo , Anilidas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Adamantano/química , Aminobenzoatos/química , Anilidas/química , Catálisis , Ciclización , Escherichia coli/genética , Escherichia coli/metabolismo , Éteres/metabolismo , Hidroxilación , Familia de Multigenes , Spirulina/genética , Spirulina/metabolismo , Estereoisomerismo
18.
Biol Pharm Bull ; 41(2): 153-157, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29386477

RESUMEN

Glucagon-like peptide-1 (GLP-1) receptor agonists (liraglutide, exenatide, lixisenatide) have recently been used as anti-diabetes drugs. We examined relationships of the binding occupancy of GLP-1 receptors (Φ) and their clinical efficacy after administration of GLP-1 receptor agonists. Next, by focusing on changes of GLP-1 concentration after administration of dipeptidyl peptidase-4 (DPP-4) inhibitors (vildagliptin, alogliptin, sitagliptin, linagliptin), we analyzed the relationship between Φ and clinical efficacy. Furthermore, using Φ as a common parameter, we compared the clinical efficacy elicited by GLP-1 receptor agonists and DPP-4 inhibitors using a theoretical analysis method. The present results showed that GLP-1 receptor agonists produced their clinical effect at a relatively low level of Φ (1.1-10.7%) at a usual dose. Furthermore, it was suggested that the drugs might achieve their full effect at an extraordinarily low level of Φ. It was also revealed that the Φ value of DPP-4 inhibitors (0.83-1.3%) was at the lower end or lower than that of GLP-1 receptor agonists at a usual dose. Accordingly, the predicted value for hemoglobin A1c (HbA1c) reduction after administration of the GLP-1 receptor agonists was higher than that of DPP-4 inhibitors. We clarified the differences between the therapeutic effects associated with GLP-1 receptor agonists and DPP-4 inhibitors theoretically. Together, the present findings provide a useful methodology for proper usage of GLP-1 receptor agonists and DPP-4 inhibitors.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Hiperglucemia/prevención & control , Hipoglucemiantes/uso terapéutico , Modelos Moleculares , Adamantano/administración & dosificación , Adamantano/análogos & derivados , Adamantano/metabolismo , Adamantano/farmacocinética , Adamantano/uso terapéutico , Algoritmos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/administración & dosificación , Inhibidores de la Dipeptidil-Peptidasa IV/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacocinética , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas , Exenatida , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hemoglobina Glucada/análisis , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacocinética , Ligandos , Liraglutida/administración & dosificación , Liraglutida/metabolismo , Liraglutida/farmacocinética , Liraglutida/uso terapéutico , Terapia Molecular Dirigida , Nitrilos/administración & dosificación , Nitrilos/metabolismo , Nitrilos/farmacocinética , Nitrilos/uso terapéutico , Péptidos/administración & dosificación , Péptidos/metabolismo , Péptidos/farmacocinética , Péptidos/uso terapéutico , Piperidinas/administración & dosificación , Piperidinas/metabolismo , Piperidinas/farmacocinética , Piperidinas/uso terapéutico , Pirrolidinas/administración & dosificación , Pirrolidinas/metabolismo , Pirrolidinas/farmacocinética , Pirrolidinas/uso terapéutico , Reproducibilidad de los Resultados , Fosfato de Sitagliptina/administración & dosificación , Fosfato de Sitagliptina/metabolismo , Fosfato de Sitagliptina/farmacocinética , Fosfato de Sitagliptina/uso terapéutico , Uracilo/administración & dosificación , Uracilo/análogos & derivados , Uracilo/metabolismo
19.
J Am Chem Soc ; 139(46): 16903-16912, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29091409

RESUMEN

5-Formylcytosine (5fC) is identified as one of the key players in active DNA demethylation and also as an epigenetic mark in mammals, thus representing a novel attractive target to chemical intervention. The current study represents an attempt to develop a reversible 5fC-targeted intervention tool. A supramolecular aldehyde reactive probe was therefore introduced for selective conversion of the 5fC to 5fC-AD nucleotide. Using various methods, we demonstrate that cucurbit[7]uril (CB7) selectively targets the 5fC-AD nucleotide in DNA, however, the binding of CB7 to 5fC-AD does not affect the hydrogen bonding properties of natural nucleobases in duplex DNA. Importantly, CB7-driven host-guest chemistry has been applied for reversible intervention of a variety of 5fC-targeted biochemical reactions, including restriction endonuclease digestion, DNA polymerase elongation, and polymerase chain reaction. On the basis of the current study, the macrocyclic CB7 creates obstructions that, through steric hindrance, prevent the enzyme from binding to the substrate, whereas the CB7/5fC-AD host-guest interactions can be reversed by treatment with adamantanamine. Moreover, fragment- and site-specific identification of 5fC modification in DNA has been accomplished without sequence restrictions. These findings thus show promising potential of host-guest chemistry for DNA/RNA epigenetics.


Asunto(s)
Adamantano/metabolismo , Aldehídos/metabolismo , Hidrocarburos Aromáticos con Puentes/metabolismo , Citosina/análogos & derivados , Imidazoles/metabolismo , Sondas Moleculares/metabolismo , Nucleósidos/metabolismo , Adamantano/química , Aldehídos/química , Hidrocarburos Aromáticos con Puentes/química , Citosina/química , Citosina/metabolismo , ADN/química , ADN/metabolismo , Imidazoles/química , Sondas Moleculares/química , Estructura Molecular , Nucleósidos/química
20.
Bioorg Med Chem Lett ; 27(4): 759-763, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28126517

RESUMEN

The adamantane scaffold, despite being widely used in medicinal chemistry, is not devoid of problems. In recent years we have developed new polycyclic scaffolds as surrogates of the adamantane group with encouraging results in multiple targets. As an adamantane scaffold is a common structural feature in several P2X7 receptor antagonists, herein we report the synthesis and pharmacological evaluation of multiple replacement options of adamantane that maintain a good activity profile. Molecular modeling studies support the binding of the compounds to a site close to the central pore, rather than to the ATP-binding site and shed light on the structural requirements for novel P2X7 antagonists.


Asunto(s)
Adamantano/química , Antagonistas del Receptor Purinérgico P2X/química , Receptores Purinérgicos P2X7/metabolismo , Adamantano/síntesis química , Adamantano/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Sitios de Unión , Células HEK293 , Humanos , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Unión Proteica , Estructura Terciaria de Proteína , Antagonistas del Receptor Purinérgico P2X/síntesis química , Antagonistas del Receptor Purinérgico P2X/metabolismo , Receptores Purinérgicos P2X7/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA