Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Eur J Med Chem ; 247: 115072, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36603510

RESUMEN

Targeting Glutathione peroxidase 4 (GPX4) has become a promising strategy for drug-resistant cancer therapy via ferroptosis induction. It was found that the GPX4 inhibitors such as RSL3 have GPX4 degradation ability via not only autophagy-lysosome pathway but also ubiquitin-proteasome system (UPS). Proteolysis targeting chimeras (PROTACs) using small molecule with both inhibition and degradation ability as the ligand of protein of interest (POI) have not been reported. To obtain better compounds with effective disturbance of GPX4 activity, and compare the difference between GPX4 inhibitors with degradation ability and their related PROTACs, we designed and synthesized a series of GPX4 degraders using PROTAC technology in terms of its excellent characteristics such as high efficiency and selectivity and the capacity of overcoming resistance. Hence, 8e was discovered as a potent and highly efficacious GPX4 degrader based upon the inhibitor RSL3. It was 2-3 times more potent than RSL3 in all the in vitro anti-tumor assays, indicating the importance of the PROTAC ternary complex of GPX4, 8e and E3 ligase ligand. 8e revealed better potency in resistant tumor cells than in wide type cells. Furthermore, we discovered for the first time that degrader 8e exhibit GPX4 degradation activity via ubiquitin-proteasome system (UPS) and autophagy-lysosome pathway with UPS plays the major role in the process. Our data also suggested that 8e and RSL3 could potently induce ferroptosis of HT1080 cells via GPX4 inhibition and degradation. In summary, our data revealed that the GPX4 degrader 8e achieves better degradation and anti-tumor effects compared to its related GPX4 inhibitor RSL3. Thus, an efficient strategy to induce GPX4 degradation and subsequent ferroptosis was established in this study for malignant cancer treatment in the future.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ligandos , Ubiquitinas/metabolismo , Proteolisis
2.
Biochem Pharmacol ; 197: 114913, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35032461

RESUMEN

Nitazoxanide and related thiazolides are a novel class of anti-infectious agents against protozoan parasites, bacteria and viruses. In recent years, it is demonstrated that thiazolides can also induce cell cycle arrest and apoptotic cell death in cancer cells. Due to their fast proliferating nature, cancer cells highly depend on the proteasome system to remove aberrant proteins. Many of these aberrant proteins are regulators of cell cycle progression and apoptosis, such as the cyclins, BCL2 family members and nuclear factor of κB (NF-κB). Here, we demonstrate at both molecular and cellular levels that the 20S proteasome is a direct target of NTZ and related thiazolides. By concurrently inhibiting the multiple catalytic subunits of 20S proteasome, NTZ promotes cell cycle arrest and triggers cell death in colon cancer cells, either directly or as a sensitizer to other anti-tumor agents, especially doxorubicin. We further show that the binding mode of NTZ in the ß5 subunit of the 20S proteasome is different from that of bortezomib and other existing proteasome inhibitors. These findings provide new insights in the design of novel small molecular proteasome inhibitors as anti-tumor agents suitable for solid tumor treatment in an oral dosing form.


Asunto(s)
Antineoplásicos/metabolismo , Muerte Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Nitrocompuestos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/metabolismo , Tiazoles/metabolismo , Animales , Antineoplásicos/administración & dosificación , Células CACO-2 , Muerte Celular/fisiología , Relación Dosis-Respuesta a Droga , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nitrocompuestos/administración & dosificación , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/administración & dosificación , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Tiazoles/administración & dosificación
3.
ACS Med Chem Lett ; 12(5): 696-703, 2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34055214

RESUMEN

We identified nitazoxanide (NTZ) as a moderate STAT3 pathway inhibitor through immunoblot analysis and a cell-based IL-6/JAK/STAT3 pathway activation assay. A series of thiazolide derivatives were designed and synthesized to further validate the thiazolide scaffold as STAT3 inhibitors. Eight out of 25 derivatives displayed potencies greater than that of NTZ, and their STAT3 pathway inhibitory activities were found to be significantly correlated with their antiproliferative activities in HeLa cells. Derivatives 15 and 24 were observed to be more potent than the positive control WP1066, which is under phase I clinical trials. Compared with NTZ, 15 also exhibited much improved in vivo pharmacokinetic parameters in rats and efficacies against proliferations in multiple cancer cell lines, indicating a broad-spectrum effect of these thiazolides as antitumor agents targeted on STAT3.

4.
ACS Med Chem Lett ; 12(5): 720-725, 2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34055217

RESUMEN

The abnormally high expression of glutathione transferases is closely associated with cancer incidence and drug resistance. By introducing a hydrophobic moiety to the inhibitor structure, we organized a series of degraders of glutathione transferases and demonstrated them potently inducing apoptosis in cancer cells, presenting their pharmacological potential in cancer therapy.

5.
ACS Sens ; 6(3): 1330-1338, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33653024

RESUMEN

In vivo imaging of cerebral hydrogen peroxide (H2O2) may facilitate early diagnosis of cerebral ischemia reperfusion injury (CIRI) and a revelation of its pathological progression. In this study, we report our rational design of a brain-targeting fluorescent probe using the basis of a pyridazinone scaffold. A structure-activity relationship study reveals that PCAB is the best candidate (Ki = 15.8 nM) for a histamine H3 receptor (H3R), which is highly expressed in neurons of the central nervous system. As a two-photon fluorescent probe, PCAB exhibits a fast, selective reaction toward both extra- and intracellular H2O2 in SH-SY5Y cells under oxygen glucose deprivation and resupply. In vivo fluorescent imaging of a middle cerebral artery occlusion mouse confirms that PCAB is an ultrasensitive probe with potent blood-brain barrier penetration, precise brain targeting, and fast detection of CIRI.


Asunto(s)
Peróxido de Hidrógeno , Daño por Reperfusión , Animales , Diagnóstico Precoz , Glucosa , Ratones , Neuronas , Daño por Reperfusión/diagnóstico
6.
Eur J Med Chem ; 218: 113386, 2021 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-33774345

RESUMEN

Mitogen-activated protein kinase kinases 1 and 2 (MEK1/2) are the crucial part of the RAS-RAF-MEK-ERK pathway (or ERK pathway), which is involved in the regulation of various cellular processes including proliferation, survival, and differentiation et al. Targeting MEK has become an important strategy for cancer therapy, and 4 MEK inhibitors (MEKis) have been approved by FDA to date. However, the application of MEKis is limited due to acquired resistance under long-term treatment. Fortunately, an emerging technology, named proteolysis targeting chimera (PROTAC), could break through this limitation by inducing MEK1/2 degradation. Compared to MEKis, MEK1/2 PROTAC is rarely studied and only three MEK1/2 PROTAC molecules, have been reported until now. This paper will outline the ERK pathway and the mechanism and research progress of MEK1/2 inhibitors, but focus on the development of MEK degraders and their optimization strategies. PAC-1 strategy which can induce MEK degradation indirectly, other PROTACs on ERK pathway, the advantages and challenges of PROTAC technology will be subsequently discussed.


Asunto(s)
Antineoplásicos/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/química , Relación Dosis-Respuesta a Droga , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Estructura Molecular , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
7.
Eur J Med Chem ; 196: 112271, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32305784

RESUMEN

A series of hybrids of MEK inhibitor and nitric oxide donor have been designed and synthesized. Compound 18h [4-(3-((3-(2-fluoro-3-((N-methylsulfamoyl)amino)benzyl)-4-methyl-2-oxo-2H-chromen-7-yl)oxy) propoxy)-3-(phenylsulfonyl)-1,2,5-oxadiazole 2-oxide] was proven to be more potent than the clinical compound RO5126766 in MDA-MB-231 cells. Compound 18h can significantly reduce the levels of pMEK and pERK, induce cell apoptosis in MDA-MB-231 cells, and release NO in cells efficiently, suggesting that these hybrids, while displaying the properties of both MEK inhibitors and NO donors have a mechanism of action different from that of MEK inhibitors and NO donors. Thus, we are able to report a series of multitarget hybrids with better antitumor potency than a known MEK inhibitor and NO donor.


Asunto(s)
Antineoplásicos/farmacología , Cumarinas/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cumarinas/síntesis química , Cumarinas/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
8.
Eur J Med Chem ; 192: 112160, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32146375

RESUMEN

A series of tripeptidic proteasome inhibitors with furylketone as C-terminus were designed and synthesized. Biochemical evaluations against ß1, ß2 and ß5 subunits revealed that they acted selectively on ß5 subunit with IC50s against chymotrypsin-like (CT-L) activity in micromolar range. LC-MS/MS analysis of the ligand-20S proteasome mixture showed that the most potent compound 11m (IC50 = 0.18 µM) made no covalent modification on 20S proteasome. However, it was identified acting in a slowly reversible manner in wash-out assay and the reversibility was much lower than that of MG132, suggesting the possibility of these tripeptidic furylketones forming reversible covalent bonds with 20S proteasome. Several compounds were selected for anti-proliferative assay towards multiple cancer cell lines, and compound 11m displayed comparable potency to positive control (MG132) in all cell lines tested. Furthermore, the pharmacokinetic (PK) data in rats indicated 11m behaved similarly (Cmax, 2007 µg/L; AUC0-t, 680 µg/L·h; Vss, 0.66 L/kg) to the clinical used agent carfilzomib. All these data suggest 11m is a good lead compound to be developed to novel anti-tumor agent.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Furanos/farmacología , Cetonas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Furanos/síntesis química , Furanos/química , Células HCT116 , Humanos , Cetonas/síntesis química , Cetonas/química , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Inhibidores de Proteasoma/síntesis química , Inhibidores de Proteasoma/química , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
9.
Anal Chem ; 92(7): 5064-5072, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32122120

RESUMEN

On the basis of the pyridazinone scaffold and photoinduced electron transfer (PET) mechanism, we designed a smart nitric oxide (NO) probe, PYSNO, with high sensitivity and selectivity. PYSNO exhibited a rapid response to both exogenous and endogenous NO. This probe can also be used in tracking and investigating NO generation in animal tissue. In the myocardial fibrosis model for mice, PYSNO exhibited a powerful imaging property in vivo as a result of unravelling the progressive relationship between the generation of myocardial NO and the occurrence of myocardial fibrosis.


Asunto(s)
Cardiomiopatías/diagnóstico por imagen , Fibrosis/diagnóstico por imagen , Colorantes Fluorescentes/química , Óxido Nítrico/análisis , Imagen Óptica , Animales , Cardiomiopatías/inducido químicamente , Células Cultivadas , Transporte de Electrón , Fibrosis/inducido químicamente , Humanos , Isoproterenol , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Estructura Molecular , Células RAW 264.7
10.
Eur J Med Chem ; 178: 802-817, 2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31252285

RESUMEN

We report in this work the discovery of novel allosteric MEK inhibitors by pharmacophore modeling and virtual screening. Two out of 13 virtual hit compounds were identified as MEK kinase inhibitors using a MEK1 binding assay. Structural derivations on the hit compound M100 (IC50 = 27.2 ±â€¯4.5 µM in RAF-MEK cascading assay) by substituent transformation and bioisosterism replacement have led to the synthesis of a small library of carbazoles. The enzymatic studies revealed the preliminary structure-activity relationships and the derivative 22k (IC50 = 12.8 ±â€¯0.5 µM) showed the most potent inhibitory effect against Raf-MEK cascading. Compound 7 was discovered as toxic as M100 to tumor cells whereas safer to HEK293 cells (IC50 > 100 µM) than M100 (IC50 = 8.9 ±â€¯2.0 µM). It suggests that carbazole is a good scaffold for the design of novel MEK inhibitors for therapeutic uses. More importantly, the developed pharmacophore model can serve as a reliable criterion in novel MEK inhibitor discovery.


Asunto(s)
Antineoplásicos/farmacología , Carbazoles/farmacología , Descubrimiento de Drogas , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Regulación Alostérica/efectos de los fármacos , Antineoplásicos/síntesis química , Antineoplásicos/química , Carbazoles/síntesis química , Carbazoles/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
11.
J Inorg Biochem ; 195: 174-181, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30954694

RESUMEN

In this report, we designed and synthesized a series of cobalt(II)-Schiff base complexes (CoSBC) with competent MEK1 (mitogen-activated protein kinase kinase-1) inhibitory activity. Based on our previous report, the CoSBC exhibited high binding affinity with MEK1 protein. To further explore metal complexes as MEK1 inhibitors, a series of transition metals and ligands were employed to build a library of various metal Schiff base complexes. The MEK inhibition assays revealed that only CoSBC exhibited obvious inhibitory activity, complex 2b showed the best inhibition both in BRaf (B-rapidly accelerated fibrosarcoma)/MEK1 and MEK1/ERK2 (extracellular signal-regulated kinases-2) cascading (IC50 is 1.988 ±â€¯0.14 µM and 1.589 ±â€¯0.054 µM respectively). In addition, homogeneous time-resolved fluorescence test method was used to prove that CoSBC as ATP-noncompetitive MEK1 inhibitor. MEK kinase selectivity assay indicated that CoSBC can selectively inhibit MEK1/2 kinases rather than other MAPKs (mitogen-activated protein kinases) family kinases. Moreover, the interaction mode of 2b with MEK1 protein has been demonstrated by computer aided drug design.


Asunto(s)
Complejos de Coordinación/química , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Bases de Schiff/química , Sitios de Unión , Complejos de Coordinación/metabolismo , Cobre/química , Diseño de Fármacos , Humanos , MAP Quinasa Quinasa 1/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Bases de Schiff/metabolismo , Relación Estructura-Actividad
12.
Chem Biol Drug Des ; 93(5): 926-933, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30667164

RESUMEN

A series of novel aminopyrimidine and diaminopyrimidine derivatives were designed and optimized to improve their potency and permeability relative to lead compound 1 (IC50  = 37.4 µM), which was discovered in a previous virtual screening. The potency of the optimized compound, 13g (IC50  = 1.4 µM), was 26-fold greater than that of 1 based on a fluorescence resonance energy transfer assay, and a parallel artificial membrane permeability assay suggested that it could pass through the blood-brain barrier. Additionally, several compounds containing selenium showed good potencies and deserve further investigation as anti-Alzheimer's agents.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores de Proteasas/síntesis química , Pirimidinas/química , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Sitios de Unión , Barrera Hematoencefálica/efectos de los fármacos , Humanos , Simulación del Acoplamiento Molecular , Permeabilidad/efectos de los fármacos , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Estructura Terciaria de Proteína , Pirimidinas/metabolismo , Pirimidinas/farmacología , Relación Estructura-Actividad
13.
Eur J Med Chem ; 137: 462-475, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28624701

RESUMEN

In this work, a series of 2-substituted-thio-N-(4-substituted-thiazol/1H-imidazol-2-yl)acetamide derivatives were developed as ß-secretase (BACE-1) inhibitors. Supported by docking study, a small library of derivatives were designed, synthesized and biologically evaluated in vitro. In addition, the selected compounds were tested with affinity (KD) towards BACE-1, blood brain barrier (BBB) permeability and cytotoxicity. The studies revealed that the most potent analog 41 (IC50 = 4.6 µM) with high predicted BBB permeability and low cellular cytotoxicity, could serve as a good lead structure for further optimization.


Asunto(s)
Acetamidas/farmacología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Acetamidas/síntesis química , Acetamidas/química , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad
14.
Bioorg Med Chem Lett ; 27(10): 2221-2224, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28385507

RESUMEN

A series of Schiff base ligands (L1-L5) and their cobalt(II) complexes (1-5) were designed and synthesized for MEK1 binding experiment. The biological evaluation results showed that Bis(N,N'-disalicylidene)-3,4-phenylenediamine-cobalt(II) 1 and Bis(N,N'-disalicylidene)-1,2-cyclohexanediamine-cobalt(II) 2 are much more effective than the parent Schiff bases (L1 and L2). Importantly, 2 exhibited MEK1 binding affinity with IC5071nM, which is so far the best result for metal complexes and more potent than U0126 (7.02µM) and AZD6244 (2.20µM). Docking study was used to elucidate the binding modes of complex 2 with MEK1. Thus cobalt(II) complex 2 may be further developed as a novel MEK1 inhibitor.


Asunto(s)
Cobalto/química , Complejos de Coordinación/química , MAP Quinasa Quinasa 1/metabolismo , Inhibidores de Proteínas Quinasas/química , Sitios de Unión , Complejos de Coordinación/síntesis química , Complejos de Coordinación/metabolismo , Ciclohexilaminas/química , Evaluación Preclínica de Medicamentos , Concentración 50 Inhibidora , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Bases de Schiff/química
15.
Bioorg Med Chem Lett ; 26(23): 5780-5784, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27816516

RESUMEN

We reported the synthesis, characterization and biological activity of several copper(II) Schiff base complexes, which exhibit high proteasome inhibitory activities with particular selectivity of ß2 subunit. Structure-activity relationships information obtained from complex Na2[Cu(a4s1)] demonstrated that distinct bonding modes in ß2 and ß5 subunits determines its selectivity and potent inhibition for ß2 subunit.


Asunto(s)
Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Cobre/química , Cobre/farmacología , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/farmacología , Diseño de Fármacos , Modelos Moleculares , Complejo de la Endopetidasa Proteasomal/metabolismo , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Bases de Schiff/química , Bases de Schiff/farmacología , Relación Estructura-Actividad
16.
Chemistry ; 22(35): 12363-70, 2016 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-27440529

RESUMEN

Phthalazinone derivatives were designed as optical probes for one- and two-photon fluorescence microscopy imaging. The design strategy involves stepwise extension and modification of pyridazinone by 1) expansion of pyridazinone to phthalazinone, a larger conjugated system, as the electron acceptor, 2) coupling of electron-donating aromatic groups such as N,N-diethylaminophenyl, thienyl, naphthyl, and quinolyl to the phthalazinone, and 3) anchoring of an alkyl chain to the phthalazinone with various terminal substituents such as triphenylphosphonio, morpholino, triethylammonio, N-methylimidazolio, pyrrolidino, and piperidino. Theoretical calculations were utilized to verify the initial design. The desired fluorescent probes were synthesized by two different routes in considerable yields. Twenty-two phthalazinone derivatives were synthesized and their photophysical properties were measured. Selected compounds were applied in cell imaging, and valuable information was obtained. Furthermore, the designed compounds showed excellent performance in two-photon microscopic imaging of mouse brain slices.


Asunto(s)
Colorantes Fluorescentes/química , Microscopía Fluorescente/métodos , Animales , Electrones , Ratones , Estructura Molecular , Fotones
17.
Bioorg Med Chem ; 24(16): 3472-82, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27288186

RESUMEN

Enterovirus 71 (EV71) is a kind of RNA virus and one of the two causes of Hand, foot and mouth disease (HFMD). Inhibitors that target key components of Ras/Raf/MEK/ERK pathway in host cells could impair replication of EV71. A series of 3-benzyl-1,3-benzoxazine-2,4-diones were designed from a specific MEK inhibitor G8935, by replacing the double bond between C3 and C4 within the coumarin scaffold with amide bond. One compound (9f) showed submicromolar inhibitory activity among the 12 derivatives. Further optimization on 9f led to two active compounds (9k and 9m) with nanomolar bioactivities (55nM and 60nM). The results of enzymatic assays also demonstrated that this series of compounds were allosteric inhibitors of unphosphorylated MEK1. The binding mode of compound 9k was predicted by molecular dynamic simulation and the key interactions were same as published MEK1/2 allosteric inhibitors. In the cell-based assays, compounds 9k and 9m could effectively suppress the ERK1/2 pathway, expression of EV71 VP1, and EV71 induced cytopathic effect (CPE) in rhabdomyosarcoma (RD) cells.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Regulación Alostérica , Antivirales/química , Línea Celular , Descubrimiento de Drogas , Humanos , Inhibidores de Proteínas Quinasas/química
18.
Bioorg Med Chem Lett ; 26(12): 2801-2805, 2016 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27158142

RESUMEN

In this study we designed a series of proteasome inhibitors using pyridazinone as initial scaffold, and extended the structure with rational design by computer aided drug design (CADD). Two different synthetic routes were explored and the biological evaluation of the phthalazinone derivatives was investigated. Most importantly, electron positive triphenylphosphine group was first introduced in the structure of proteasome inhibitors and potent inhibition was achieved. As 6c was the most potent inhibitor of proteasome, we examined the structure-activity relationship (SAR) of 6c analogs.


Asunto(s)
Ftalazinas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Saccharomyces cerevisiae/enzimología , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Ftalazinas/síntesis química , Ftalazinas/química , Inhibidores de Proteasoma/síntesis química , Inhibidores de Proteasoma/química , Relación Estructura-Actividad
19.
ChemMedChem ; 10(3): 498-510, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25641789

RESUMEN

Proteasome inhibitors are promising compounds for a number of therapies, including cardiovascular and eye diseases, diabetes, and cancers. We previously reported a series of furan-based peptidic inhibitors with moderate potencies against the proteasome ß5 subunit, hypothesizing that the C-terminal furyl ketone motif could form a covalent bond with the catalytic residue, threonine 1. In this context, we describe further optimizations of the furan-based peptides, and a series of dipeptidic and tripeptidic inhibitors were designed and synthesized, aiming at improved potency and better solubility. Most of the tripeptidic inhibitors demonstrated improved potency and selectivity as ß5 subunit inhibitors in both enzymatic and cellular assays, and good antineoplastic activities in various tumor cell lines were also observed. However, no inhibitory effects were observed for the dipeptidic compounds, which led us to presume that a noncovalent binding mode is adopted. Docking studies and molecular dynamics simulations were carried out to verify this presumption, with results showing that the distance between the furyl ketone motif and Thr1 is slightly too long to form covalent bond.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Furanos/química , Furanos/farmacología , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/farmacología , Antineoplásicos/síntesis química , Línea Celular Tumoral , Furanos/síntesis química , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Peptidomiméticos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/síntesis química , Unión Proteica
20.
Eur J Med Chem ; 79: 413-21, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24763262

RESUMEN

In this work, we report a series of new 4-oxo-1,4-dihydro-quinoline-3-carboxamide derivatives as ß-secretase (BACE-1) inhibitors. Supported by docking study, a small library of derivatives were designed, synthesized and biologically evaluated in vitro. The studies revealed that the most potent analog 14e (IC50 = 1.89 µM) with low cellular cytotoxicity and high predicted blood brain barrier permeability, could serve as a good structure for further modification.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Quinolinas/farmacología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Modelos Moleculares , Estructura Molecular , Quinolinas/síntesis química , Quinolinas/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA