Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Chem Sci ; 12(34): 11484-11489, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34667552

RESUMO

Rapadocin is a novel rapamycin-inspired polyketide-tetrapeptide hybrid macrocycle that possesses highly potent and isoform-specific inhibitory activity against the human equilibrative nucleoside transporter 1 (hENT1). Rapadocin contains an epimerizable chiral center in phenylglycine and an olefin group, and can thus exist as a mixture of four stereoisomers. Herein, we report the first total synthesis of the four stereoisomers of rapadocin using two different synthetic strategies and the assignment of their structures. The inhibitory activity of each of the four synthetic isomers on both hENT1 and hENT2 was determined. It was found that the stereochemistry of phenylglycine played a more dominant role than the configuration of the olefin in the activity of rapadocin. These findings will guide the future design and development of rapadocin analogs as new modulators of adenosine signaling.

2.
Angew Chem Int Ed Engl ; 58(48): 17158-17162, 2019 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-31591797

RESUMO

Glucose transporters play an essential role in cancer cell proliferation and survival and have been pursued as promising cancer drug targets. Using microarrays of a library of new macrocycles known as rapafucins, which were inspired by the natural product rapamycin, we screened for new inhibitors of GLUT1. We identified multiple hits from the rapafucin 3D microarray and confirmed one hit as a bona fide GLUT1 ligand, which we named rapaglutin A (RgA). We demonstrate that RgA is a potent inhibitor of GLUT1 as well as GLUT3 and GLUT4, with an IC50 value of low nanomolar for GLUT1. RgA was found to inhibit glucose uptake, leading to a decrease in cellular ATP synthesis, activation of AMP-dependent kinase, inhibition of mTOR signaling, and induction of cell-cycle arrest and apoptosis in cancer cells. Moreover, RgA was capable of inhibiting tumor xenografts in vivo without obvious side effects. RgA could thus be a new chemical tool to study GLUT function and a promising lead for developing anticancer drugs.


Assuntos
Antineoplásicos/química , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Macrolídeos/farmacologia , Bibliotecas de Moléculas Pequenas/química , Células A549 , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Células MCF-7 , Macrolídeos/química , Estrutura Molecular , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Análise Serial de Proteínas , Transdução de Sinais , Sirolimo/química , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/metabolismo , Tacrolimo/química , Proteínas de Ligação a Tacrolimo
3.
ACS Chem Biol ; 12(1): 174-182, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-28103683

RESUMO

The antifungal drug itraconazole was recently found to exhibit potent antiangiogenic activity and has since been repurposed as an investigational anticancer agent. Itraconazole has been shown to exert its antiangiogenic activity through inhibition of the mTOR signaling pathway, but the molecular mechanism of action was unknown. We recently identified the mitochondrial protein VDAC1 as a target of itraconazole and a mediator of its activation of AMPK, an upstream regulator of mTOR. However, VDAC1 could not account for the previously reported inhibition of cholesterol trafficking by itraconazole, which was also demonstrated to lead to mTOR inhibition. In this study, we demonstrate that cholesterol trafficking inhibition by itraconazole is due to direct inhibition of the lysosomal protein NPC1. We further map the binding site of itraconazole to the sterol-sensing domain of NPC1 using mutagenesis, competition with U18666A, and molecular docking. Finally, we demonstrate that simultaneous AMPK activation and cholesterol trafficking inhibition leads to synergistic inhibition of mTOR, endothelial cell proliferation, and angiogenesis.


Assuntos
Inibidores da Angiogênese/farmacologia , Antifúngicos/farmacologia , Proteínas de Transporte/metabolismo , Itraconazol/farmacologia , Glicoproteínas de Membrana/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Transporte Biológico/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colesterol/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Simulação de Acoplamento Molecular , Proteína C1 de Niemann-Pick , Serina-Treonina Quinases TOR/antagonistas & inibidores
4.
J Med Chem ; 54(22): 7751-8, 2011 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-22003962

RESUMO

Improved therapies are needed for nonsmall cell lung cancer. Diazeniumdiolate-based nitric oxide (NO)-releasing prodrugs are a growing class of promising NO-based therapeutics. Recently, we have shown that O(2)-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate (JS-K, 1) is effective against nonsmall cell lung cancer (NSCLC) cells in culture and in vivo. Here we report mechanistic studies with compound 1 and its homopiperazine analogue and structural modification of these into more stable prodrugs. Compound 1 and its homopiperazine analogue were potent cytotoxic agents against NSCLC cells in vitro and in vivo, concomitant with activation of the SAPK/JNK stress pathway and upregulation of its downstream effector ATF3. Apoptosis followed these events. An aryl-substituted analogue, despite extended half-life in the presence of glutathione, did not activate JNK or have antitumor activity. The data suggest that rate of reactivity with glutathione and activation of JNK/ATF3 are determinants of cancer cell killing by these prodrugs.


Assuntos
Fator 3 Ativador da Transcrição/fisiologia , Antineoplásicos/síntese química , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Doadores de Óxido Nítrico/síntese química , Pró-Fármacos/síntese química , Fator 3 Ativador da Transcrição/biossíntese , Fator 3 Ativador da Transcrição/genética , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Pontos de Checagem do Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Ativação Enzimática , Fase G2 , Inativação Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Doadores de Óxido Nítrico/química , Doadores de Óxido Nítrico/farmacologia , Piperazinas/síntese química , Piperazinas/química , Piperazinas/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Transdução de Sinais , Relação Estrutura-Atividade , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Chem Biol Interact ; 193(1): 88-96, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21621526

RESUMO

Arsenic is a cancer chemotherapeutic but hepatotoxicity can be a limiting side effect. O(2)-vinyl 1-[2-(carboxylato)pyrrolidin-1-yl]diazen-1-ium-1,2-diolate (V-PROLI/NO) is a nitric oxide (NO) donor prodrug and metabolized by liver cytochromes P450 (CYP450) to release NO. The effects of V-PROLI/NO pretreatment on the toxicity of arsenic (as NaAsO(2)) were studied in a rat liver cell line (TRL 1215). The cells acted upon the prodrug to release NO, as assessed by nitrite levels, in a time-dependent fashion to maximal levels of 8-fold above basal levels. Pretreatment with V-PROLI/NO markedly reduced arsenic cytolethality which was directly related to the level of NO produced by V-PROLI/NO treatment. Cyp1a1 expression was directly related to the level of NO production and to reduced arsenic cytotoxicity. V-PROLI/NO pretreatment markedly reduced arsenic-induced apoptosis and suppressed phosphorylation of JNK1/2. V-PROLI/NO pretreatment facilitated additional increases in arsenic-induced metallothionein, a metal-binding protein important in arsenic tolerance. Thus, V-PROLI/NO protects against arsenic toxicity in rat liver cells, reducing cytolethality, apoptosis and dysregulation of MAPKs, through generation of NO formed after metabolism by liver cell enzymes, possibly including Cyp1a1. CYP450 required for NO production from V-PROLI/NO treatment in the rat and human appears to differ as we have previously studied the ability of V-PROLI/NO to prevent arsenic toxicity in human liver cells where it reduced toxicity apparently through a CYP2E1-mediated metabolic mechanism. None-the-less, it appears that both rat and human liver cells act upon V-PROLI/NO via a CYP450-related mechanism to produce NO and subsequently reduce arsenic toxicity.


Assuntos
Arsênio/toxicidade , Citocromo P-450 CYP1A1/metabolismo , Hepatócitos/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Pró-Fármacos/farmacologia , Pirrolidinas/farmacologia , Triazenos/farmacologia , Animais , Apoptose , Linhagem Celular , Citocromo P-450 CYP1A1/genética , Hepatócitos/metabolismo , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase 7/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/química , Pró-Fármacos/química , Pirrolidinas/química , Ratos , Triazenos/química
6.
Mol Pharm ; 7(1): 291-8, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20000791

RESUMO

We report the stabilization of the nitric oxide (NO) prodrugs and anticancer lead compounds, PABA/NO (O(2)-{2,4-dinitro-5-[4-(N-methylamino)benzoyloxy]phenyl} 1-(N,N-dimethylamino)diazen-1-ium-1,2-diolate) and "Double JS-K" 1,5-bis-{1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diol-2-ato}-2,4-dinitrobenzene, through their incorporation into polymer-protected nanoparticles. The prodrugs were formulated in block copolymer-stabilized nanoparticles with sizes from 220 to 450 nm by a novel rapid precipitation process. The block copolymers, with polyethylene glycol (PEG) soluble blocks, provide a steric barrier against NO prodrug activation by glutathione. Too rapid activation and NO release has been a major barrier to effective administration of this class of compounds. The nanoparticle stabilized PABA/NO are protected from attack by glutathione as evidenced by a significant increase in time taken for 50% decomposition from 15 min (unformulated) to 5 h (formulated); in the case of Double JS-K, the 50% decomposition time was extended from 4.5 min (unformulated) to 40 min (formulated). The more hydrophobic PABA/NO produced more stable nanoparticles and correspondingly more extended release times in comparison with Double JS-K. The hydrophobic blocks of the polymer were either polystyrene or polylactide. Both blocks produced nanoparticles of approximately the same size and release kinetics. This combination of PEG-protected nanoparticles with sizes appropriate for cancer targeting by enhanced permeation and retention (EPR) and delayed release of NO may afford enhanced therapeutic benefit.


Assuntos
Antineoplásicos/administração & dosagem , Doadores de Óxido Nítrico/administração & dosagem , Pró-Fármacos/administração & dosagem , Ácido 4-Aminobenzoico/administração & dosagem , Ácido 4-Aminobenzoico/química , Compostos Azo/administração & dosagem , Compostos Azo/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Preparações de Ação Retardada , Estabilidade de Medicamentos , Humanos , Nanopartículas , Doadores de Óxido Nítrico/química , Tamanho da Partícula , Piperazinas/administração & dosagem , Piperazinas/química , Polietilenoglicóis , Pró-Fármacos/química , para-Aminobenzoatos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA