Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Biol Chem ; 289(11): 7825-34, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24500713

RESUMO

ARTEMIS is a member of the metallo-ß-lactamase protein family. ARTEMIS has endonuclease activity at DNA hairpins and at 5'- and 3'-DNA overhangs of duplex DNA, and this endonucleolytic activity is dependent upon DNA-PKcs. There has been uncertainty about whether ARTEMIS also has 5'-exonuclease activity on single-stranded DNA and 5'-overhangs, because this 5'-exonuclease is not dependent upon DNA-PKcs. Here, we show that the 5'-exonuclease and the endonuclease activities co-purify. Second, we show that a point mutant of ARTEMIS at a putative active site residue (H115A) markedly reduces both the endonuclease activity and the 5'-exonuclease activity. Third, divalent cation effects on the 5'-exonuclease and the endonuclease parallel one another. Fourth, both the endonuclease activity and 5'-exonuclease activity of ARTEMIS can be blocked in parallel by small molecule inhibitors, which do not block unrelated nucleases. We conclude that the 5'-exonuclease is intrinsic to ARTEMIS, making it relevant to the role of ARTEMIS in nonhomologous DNA end joining.


Assuntos
DNA/química , Desoxirribonuclease I/metabolismo , Exodesoxirribonucleases/metabolismo , Proteínas Nucleares/metabolismo , Nucleotidases/química , Cromatografia , Dicroísmo Circular , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA , Endonucleases , Células HEK293 , Humanos , Mutagênese , Proteínas Nucleares/genética , Oligonucleotídeos/química , Mutação Puntual , Transfecção
2.
Bioorg Med Chem Lett ; 22(2): 1237-41, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22182498

RESUMO

The overproduction of nitric oxide during the biological response to inflammation by the nitric oxide synthase (NOS) enzymes have been implicated in the pathology of many diseases. By removal of the amide core from uHTS-derived quinolone 4, a new series highly potent heteroaromatic-aminomethyl quinolone iNOS inhibitors 8 were identified. SAR studies led to identification of piperazine 22 and pyrimidine 32, both of which reduced plasma nitrates following oral dosing in a mouse lipopolysaccharide challenge assay.


Assuntos
Inibidores Enzimáticos/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Quinolonas/farmacologia , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular , Óxido Nítrico Sintase Tipo II/metabolismo , Quinolonas/síntese química , Quinolonas/química , Estereoisomerismo , Relação Estrutura-Atividade
3.
J Pharmacol Exp Ther ; 336(2): 468-78, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21036913

RESUMO

Nitric oxide (NO) derived from neuronal nitric-oxide synthase (nNOS) and inducible nitric-oxide synthase (iNOS) plays a key role in various pain and inflammatory states. KLYP961 (4-((2-cyclobutyl-1H-imidazo[4,5-b]pyrazin-1-yl)methyl)-7,8-difluoroquinolin-2(1H)-one) inhibits the dimerization, and hence the enzymatic activity of human, primate, and murine iNOS and nNOS (IC(50) values 50-400 nM), with marked selectivity against endothelial nitric-oxide synthase (IC(50) >15,000 nM). It has ideal drug like-properties, including excellent rodent and primate pharmacokinetics coupled with a minimal off-target activity profile. In mice, KLYP961 attenuated endotoxin-evoked increases in plasma nitrates, a surrogate marker of iNOS activity in vivo, in a sustained manner (ED(50) 1 mg/kg p.o.). KLYP961 attenuated pain behaviors in a mouse formalin model (ED(50) 13 mg/kg p.o.), cold allodynia in the chronic constriction injury model (ED(50) 25 mg/kg p.o.), or tactile allodynia in the spinal nerve ligation model (ED(50) 30 mg/kg p.o.) with similar efficacy, but superior potency relative to gabapentin, pregabalin, or duloxetine. Unlike morphine, the antiallodynic activity of KLYP961 did not diminish upon repeated dosing. KLYP961 also attenuated carrageenin-induced edema and inflammatory hyperalgesia and writhing response elicited by phenylbenzoquinone with efficacy and potency similar to those of celecoxib. In contrast to gabapentin, KLYP961 did not impair motor coordination at doses as high as 1000 mg/kg p.o. KLYP961 also attenuated capsaicin-induced thermal allodynia in rhesus primates in a dose-related manner with a minimal effective dose (≤ 10 mg/kg p.o.) and a greater potency than gabapentin. In summary, KLYP961 represents an ideal tool with which to probe the physiological role of NO derived from iNOS and nNOS in human pain and inflammatory states.


Assuntos
Anti-Inflamatórios/farmacologia , Inibidores Enzimáticos/farmacologia , Fluoroquinolonas/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Pirazinas/farmacologia , Analgésicos/farmacologia , Animais , Células Cultivadas , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/toxicidade , Fluoroquinolonas/farmacocinética , Fluoroquinolonas/toxicidade , Trânsito Gastrointestinal/efeitos dos fármacos , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Multimerização Proteica , Pirazinas/farmacocinética , Pirazinas/toxicidade
4.
Bioorg Med Chem Lett ; 21(22): 6888-94, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21986586

RESUMO

We have identified and synthesized a series of imidazole containing dimerization inhibitors of inducible nitric oxide synthase (iNOS). The necessity of key imidazole and piperonyl functionality was demonstrated and SAR studies led to the identification of compound 35, which showed a dose dependant inhibition in multiple pain models, including tactile allodynia induced by spinal nerve ligation (Chung model).


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Imidazóis/química , Imidazóis/uso terapêutico , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Dor/tratamento farmacológico , Multimerização Proteica/efeitos dos fármacos , Animais , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Imidazóis/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Endogâmicos Lew
5.
Cancer Res ; 81(6): 1472-1485, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33414172

RESUMO

Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. SIGNIFICANCE: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Endossomos/metabolismo , Invasividade Neoplásica/patologia , Podossomos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Dineínas do Citoplasma/genética , Dineínas do Citoplasma/metabolismo , Conjuntos de Dados como Assunto , Matriz Extracelular , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Invasividade Neoplásica/prevenção & controle , Podossomos/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Imagem com Lapso de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Pharmacol ; 76(1): 153-62, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19364813

RESUMO

Nitric-oxide synthases (NOS) generate nitric oxide (NO) through the oxidation of l-arginine. Inappropriate or excessive production of NO by NOS is associated with the pathophysiology of various disease states. Efforts to treat these disorders by developing arginine mimetic, substrate-competitive NOS inhibitors as drugs have met with little success. Small-molecule-mediated inhibition of NOS dimerization represents an intriguing alternative to substrate-competitive inhibition. An ultra-high-throughput cell-based screen of 880,000 small molecules identified a novel quinolinone with inducible NOS (iNOS) inhibitory activity. Exploratory chemistry based on this initial screening hit resulted in the synthesis of KLYP956, which inhibits iNOS at low nanomolar concentrations. The iNOS inhibitory potency of KLYP956 is insensitive to changes in concentrations of the substrate arginine, or the cofactor tetrahydrobiopterin. Mechanistic analysis suggests that KLYP956 binds the oxygenase domain in the vicinity of the active site heme and inhibits iNOS and neuronal NOS (nNOS) by preventing the formation of enzymatically active dimers. Oral administration of KLYP956 [N-(3-chlorophenyl)-N-((8-fluoro-2-oxo-1,2-dihydroquinolin-4-yl)methyl)-4-methylthiazole-5-carboxamide] inhibits iNOS activity in a murine model of endotoxemia and blocks pain behaviors in a formalin model of nociception. KLYP956 thus represents the first nonimidazole-based inhibitor of iNOS and nNOS dimerization and provides a novel pharmaceutical alternative to previously described substrate competitive inhibitors.


Assuntos
Inibidores Enzimáticos/farmacologia , Fluoroquinolonas/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Tiazóis/farmacologia , Administração Oral , Animais , Células Cultivadas , Dimerização , Humanos , Camundongos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/química , Óxido Nítrico Sintase Tipo I/química , Óxido Nítrico Sintase Tipo II/química , Dor/tratamento farmacológico , Especificidade da Espécie
7.
Mol Cancer Ther ; 7(6): 1494-505, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18566220

RESUMO

Histone deacetylase inhibitors have emerged as promising anticancer drugs. Using an unbiased ultrahigh throughput screening system, a novel mercaptoketone-based histone deacetylase inhibitor series was identified that was optimized to the lead compound, KD5170. KD5170 inhibited the proliferation of myeloma cell lines and the viability of CD138(+) primary myeloma cells by induction of apoptosis, accompanied by an increase of acetylation of histones and activation of caspase-3, caspase-8, and caspase-9. Treatment with KD5170 caused a loss of mitochondrial membrane potential resulting in release of apoptogenic factors such as cytochrome c, Smac, and apoptosis-inducing factor. Furthermore, KD5170 induced oxidative stress and oxidative DNA damage in myeloma cells as evidenced by the up-regulation of heme oxygenase-1 and H2A.X phosphorylation. Combination of KD5170 with proteasome inhibitor bortezomib or tumor necrosis factor-related apoptosis-inducing ligand synergistically enhanced the antimyeloma activity. We further found that resistance of myeloma cells to KD5170 was associated with activation of the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway under treatment with KD5170. Pretreatment with the mitogen-activated protein kinase inhibitor U0126 restored sensitivity to KD5170, suggesting that the combination of KD5170 with U0126 could overcome drug resistance. Growth of myeloma tumor xenografts in KD5170-treated nude mice was significantly inhibited and survival was prolonged. Histone acetylation was increased in spleen and tumor tissues of animals treated with KD5170. Our data indicate that KD5170 has potent antimyeloma activity in vitro and in vivo, which is mediated by DNA damage and mitochondrial signaling and subsequent induction of apoptosis.


Assuntos
Dano ao DNA , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Cetonas/farmacologia , Mitocôndrias/metabolismo , Mieloma Múltiplo/enzimologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Acetilação/efeitos dos fármacos , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Ácidos Borônicos/farmacologia , Bortezomib , Caspases/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/química , Histonas/metabolismo , Humanos , Cetonas/química , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Pirazinas/farmacologia , Piridinas/química , Sulfonamidas/química , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2/metabolismo
8.
Mol Cancer Ther ; 7(5): 1054-65, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18483295

RESUMO

Histone deacetylase (HDAC) inhibitors have garnered significant attention as cancer drugs. These therapeutic agents have recently been clinically validated with the market approval of vorinostat (SAHA, Zolinza) for treatment of cutaneous T-cell lymphoma. Like vorinostat, most of the small-molecule HDAC inhibitors in clinical development are hydroxamic acids, whose inhibitory activity stems from their ability to coordinate the catalytic Zn2+ in the active site of HDACs. We sought to identify novel, nonhydroxamate-based HDAC inhibitors with potentially distinct pharmaceutical properties via an ultra-high throughput small molecule biochemical screen against the HDAC activity in a HeLa cell nuclear extract. An alpha-mercaptoketone series was identified and chemically optimized. The lead compound, KD5170, exhibits HDAC inhibitory activity with an IC50 of 0.045 micromol/L in the screening biochemical assay and an EC50 of 0.025 micromol/L in HeLa cell-based assays that monitor histone H3 acetylation. KD5170 also exhibits broad spectrum classes I and II HDAC inhibition in assays using purified recombinant human isoforms. KD5170 shows significant antiproliferative activity against a variety of human tumor cell lines, including the NCI-60 panel. Significant tumor growth inhibition was observed after p.o. dosing in human HCT-116 (colorectal cancer), NCI-H460 (non-small cell lung carcinoma), and PC-3 (prostate cancer) s.c. xenografts in nude mice. In addition, a significant increase in antitumor activity and time to end-point occurred when KD5170 was combined with docetaxel in xenografts of the PC-3 prostate cancer cell line. The biological and pharmaceutical profile of KD5170 supports its continued preclinical and clinical development as a broad spectrum anticancer agent.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Piridinas/farmacologia , Sulfonamidas/farmacologia , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Thorac Oncol ; 14(12): 2152-2163, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31470128

RESUMO

INTRODUCTION: Despite the enthusiasm surrounding cancer immunotherapy, most SCLC patients show very modest response to immune checkpoint inhibitor monotherapy treatment. Therefore, there is growing interest in combining immune checkpoint blockade with chemotherapy and other treatments to enhance immune checkpoint blockade efficacy. Based on favorable clinical trial results, chemotherapy and immunotherapy combinations have been recently approved by the U.S. Food and Drug Administration for frontline treatment for SCLC. METHODS AND RESULTS: Here, we show that combined treatment of SRA737, an oral CHK1 inhibitor, and anti-programmed death ligand 1 (PD-L1) leads to an antitumor response in multiple cancer models, including SCLC. We further show that combining low, non-cytotoxic doses of gemcitabine with SRA737 + anti-PD-L1/anti-PD-1 significantly increased antitumorigenic CD8+ cytotoxic T cells, dendritic cells, and M1 macrophage populations in an SCLC model. This regimen also led to a significant decrease in immunosuppressive M2 macrophage and myeloid-derived suppressor cell populations, as well as an increase in the expression of the type I interferon beta 1 gene, IFNß, and chemokines, CCL5 and CXCL10. CONCLUSIONS: Given that anti-PD-L1/anti-PD-1 drugs have recently been approved as monotherapy and in combination with chemotherapy for the treatment of SCLC, and that the SRA737 + low dose gemcitabine regimen is currently in clinical trials for SCLC and other malignancies, our preclinical data provide a strong rational for combining this regimen with inhibitors of the PD-L1/PD-1 pathway.


Assuntos
Terapia Combinada/métodos , Desoxicitidina/análogos & derivados , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Imunoterapia/métodos , Neoplasias Pulmonares/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Microambiente Tumoral/imunologia , Administração Oral , Animais , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Carcinoma de Pequenas Células do Pulmão/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
10.
Bioorg Med Chem Lett ; 18(23): 6093-6, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18954983

RESUMO

We report the identification of KD5170, a potent mercaptoketone-based Class I and II-histone deacetylase inhibitor that demonstrates broad spectrum cytotoxic activity against a range of human tumor-derived cell lines. KD5170 exhibits robust and sustained histone H3 hyperacetylation in HCT-116 xenograft tumors following single oral or i.v. dose and inhibition of tumor growth following chronic dosing.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Pró-Fármacos/farmacologia , Piridinas/farmacologia , Sulfonamidas/farmacologia , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Células HCT116 , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Nus , Estrutura Molecular , Pró-Fármacos/química , Piridinas/química , Relação Estrutura-Atividade , Sulfonamidas/química , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Bioorg Med Chem Lett ; 18(24): 6482-5, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18954984

RESUMO

In an effort to discover novel non-hydroxamic acid histone deacetylase (HDAC) inhibitors, a novel alpha-mercaptoketone was identified in a high-throughput screen. Lead optimization of the screening hit, led to a number of potent HDAC inhibitors. In particular, alpha-mercaptoketone 19y (KD5150) exhibited nanomolar in vitro activity and inhibition of tumor growth in vivo.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais , Inibidores de Histona Desacetilases , Cetonas/química , Antineoplásicos/uso terapêutico , Quelantes/farmacologia , Química Farmacêutica , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Modelos Químicos , Neoplasias/tratamento farmacológico , Pró-Fármacos/química , Relação Estrutura-Atividade , Zinco/química
12.
PLoS One ; 11(3): e0152692, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27031987

RESUMO

Cardiac glycosides (CGs), inhibitors of Na+/K+-ATPase (NKA), used clinically to treat heart failure, have garnered recent attention as potential anti-cancer and anti-viral agents. A high-throughput phenotypic screen designed to identify modulators of promyelocytic leukemia protein (PML) nuclear body (NB) formation revealed the CG gitoxigenin as a potent activator of PML. We demonstrate that multiple structurally distinct CGs activate the formation of PML NBs and induce PML protein SUMOylation in an NKA-dependent fashion. CG effects on PML occur at the post-transcriptional level, mechanistically distinct from previously described PML activators and are mediated through signaling events downstream of NKA. Curiously, genomic deletion of PML in human cancer cells failed to abrogate the cytotoxic effects of CGs and other apoptotic stimuli such as ceramide and arsenic trioxide that were previously shown to function through PML in mice. These findings suggest that alternative pathways can compensate for PML loss to mediate apoptosis in response to CGs and other apoptotic stimuli.


Assuntos
Glicosídeos Cardíacos/farmacologia , Proteínas Nucleares/metabolismo , Sumoilação/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose/efeitos dos fármacos , Glicosídeos Cardíacos/química , Chlorocebus aethiops , Deleção de Genes , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Células Vero
13.
J Biomol Screen ; 20(10): 1232-45, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26265713

RESUMO

Endoplasmic reticulum (ER) stress activates three distinct signal transducers on the ER membrane. Inositol-requiring protein 1 (IRE1), the most conserved signal transducer, plays a key role in ER stress-mediated signaling. During ER stress, IRE1 initiates two discrete signaling cascades: the "adaptive" signaling cascade mediated by the XBP1 pathway and the "alarm" signaling cascade mediated by stress-activated protein kinase pathways. Fine-tuning of the balance between the adaptive and alarm signals contributes significantly to cellular fate under ER stress. Thus, we propose that the design of high-throughput screening (HTS) assays to selectively monitor IRE1 mediated-signaling would be desirable for drug discovery. To this end, we report the generation of stable human neural cell lines and development of cell-based HTS luciferase (Luc) reporter gene assays for the identification of pathway-specific chemical modulators of IRE1. We implemented a cell-based Luc assay using a chimeric CHOP-Gal4 transcription factor in 384-well format for monitoring IRE1 kinase-mediated p38MAPK activation and an unfolded response pathway element (URPE)-Luc cell-based assay in 1536-well format for monitoring IRE1's RNase-mediated activation of XBP1. Chemical library screening was successfully conducted with both the CHOP/Gal4-Luc cells and UPRE-Luc engineered cells. The studies demonstrate the feasibility of using these HTS assays for discovery of pathway-selective modulators of IRE1.


Assuntos
Endorribonucleases/antagonistas & inibidores , Ensaios de Triagem em Larga Escala , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Estresse do Retículo Endoplasmático , Endorribonucleases/fisiologia , Ativação Enzimática , Genes Reporter , Células HeLa , Humanos , Luciferases/análise , Luciferases/genética , Sistema de Sinalização das MAP Quinases , Neurônios , Proteínas Serina-Treonina Quinases/fisiologia , Fatores de Transcrição de Fator Regulador X , Tapsigargina/metabolismo , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a X-Box
14.
Cancer Res ; 75(24): 5211-8, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26603897

RESUMO

Disrupting the eukaryotic translation initiation factor 4F (eIF4F) complex offers an appealing strategy to potentiate the effectiveness of existing cancer therapies and to overcome resistance to drugs such as BRAF inhibitors (BRAFi). Here, we identified and characterized the small molecule SBI-0640756 (SBI-756), a first-in-class inhibitor that targets eIF4G1 and disrupts the eIF4F complex. SBI-756 impaired the eIF4F complex assembly independently of mTOR and attenuated growth of BRAF-resistant and BRAF-independent melanomas. SBI-756 also suppressed AKT and NF-κB signaling, but small-molecule derivatives were identified that only marginally affected these pathways while still inhibiting eIF4F complex formation and melanoma growth, illustrating the potential for further structural and functional manipulation of SBI-756 as a drug lead. In the gene expression signature patterns elicited by SBI-756, DNA damage, and cell-cycle regulatory factors were prominent, with mutations in melanoma cells affecting these pathways conferring drug resistance. SBI-756 inhibited the growth of NRAS, BRAF, and NF1-mutant melanomas in vitro and delayed the onset and reduced the incidence of Nras/Ink4a melanomas in vivo. Furthermore, combining SBI-756 and a BRAFi attenuated the formation of BRAFi-resistant human tumors. Taken together, our findings show how SBI-756 abrogates the growth of BRAF-independent and BRAFi-resistant melanomas, offering a preclinical rationale to evaluate its antitumor effects in other cancers.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Fator de Iniciação 4F em Eucariotos/metabolismo , Lactamas/farmacologia , Melanoma/patologia , Quinolonas/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Humanos , Melanoma/metabolismo , Camundongos
15.
J Biomol Screen ; 19(8): 1201-11, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24870016

RESUMO

Antiapoptotic Bcl-2 family proteins are validated cancer targets composed of six related proteins. From a drug discovery perspective, these are challenging targets that exert their cellular functions through protein-protein interactions (PPIs). Although several isoform-selective inhibitors have been developed using structure-based design or high-throughput screening (HTS) of synthetic chemical libraries, no large-scale screen of natural product collections has been reported. A competitive displacement fluorescence polarization (FP) screen of nearly 150,000 natural product extracts was conducted against all six antiapoptotic Bcl-2 family proteins using fluorochrome-conjugated peptide ligands that mimic functionally relevant PPIs. The screens were conducted in 1536-well format and displayed satisfactory overall HTS statistics, with Z'-factor values ranging from 0.72 to 0.83 and a hit confirmation rate between 16% and 64%. Confirmed active extracts were orthogonally tested in a luminescent assay for caspase-3/7 activation in tumor cells. Active extracts were resupplied, and effort toward the isolation of pure active components was initiated through iterative bioassay-guided fractionation. Several previously described altertoxins were isolated from a microbial source, and the pure compounds demonstrate activity in both Bcl-2 FP and caspase cellular assays. The studies demonstrate the feasibility of ultra-high-throughput screening using natural product sources and highlight some of the challenges associated with this approach.


Assuntos
Produtos Biológicos/química , Ensaios de Triagem em Larga Escala/métodos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Células CACO-2 , Caspase 3/metabolismo , Caspase 7/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Polarização de Fluorescência/métodos , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Miniaturização , Terapia de Alvo Molecular/métodos , Micotoxinas/isolamento & purificação , Micotoxinas/farmacologia , Extração em Fase Sólida , Proteína bcl-X/antagonistas & inibidores
16.
Clin Cancer Res ; 19(10): 2710-22, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23589174

RESUMO

PURPOSE: Effective therapy for malignant melanoma, the leading cause of death from skin cancer, remains an area of significant unmet need in oncology. The elevated expression of PKCε in advanced metastatic melanoma results in the increased phosphorylation of the transcription factor ATF2 on threonine 52, which causes its nuclear localization and confers its oncogenic activities. The nuclear-to-mitochondrial translocation of ATF2 following genotoxic stress promotes apoptosis, a function that is largely lost in melanoma cells, due to its confined nuclear localization. Therefore, promoting the nuclear export of ATF2, which sensitizes melanoma cells to apoptosis, represents a novel therapeutic modality. EXPERIMENTAL DESIGN: We conducted a pilot high-throughput screen of 3,800 compounds to identify small molecules that promote melanoma cell death by inducing the cytoplasmic localization of ATF2. The imaging-based ATF2 translocation assay was conducted using UACC903 melanoma cells that stably express doxycycline-inducible GFP-ATF2. RESULTS: We identified two compounds (SBI-0089410 and SBI-0087702) that promoted the cytoplasmic localization of ATF2, reduced cell viability, inhibited colony formation, cell motility, and anchorage-free growth, and increased mitochondrial membrane permeability. SBI-0089410 inhibited the 12-O-tetradecanoylphorbol-l3-acetate (TPA)-induced membrane translocation of protein kinase C (PKC) isoforms, whereas both compounds decreased ATF2 phosphorylation by PKCε and ATF2 transcriptional activity. Overexpression of either constitutively active PKCε or phosphomimic mutant ATF2(T52E) attenuated the cellular effects of the compounds. CONCLUSION: The imaging-based high-throughput screen provides a proof-of-concept for the identification of small molecules that block the oncogenic addiction to PKCε signaling by promoting ATF2 nuclear export, resulting in mitochondrial membrane leakage and melanoma cell death.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fator 2 Ativador da Transcrição/genética , Animais , Antineoplásicos/química , Benzamidas/química , Benzamidas/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Immunoblotting , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Microscopia Confocal , Mitocôndrias/metabolismo , Estrutura Molecular , Células NIH 3T3 , Naftalenos/química , Naftalenos/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenetilaminas/química , Fenetilaminas/farmacologia , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , Transporte Proteico/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Sulfonamidas/química , Sulfonamidas/farmacologia
17.
J Med Chem ; 53(21): 7739-55, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20931971

RESUMO

Three isoforms of nitric oxide synthase (NOS), dimeric enzymes that catalyze the formation of nitric oxide (NO) from arginine, have been identified. Inappropriate or excessive NO produced by iNOS and/or nNOS is associated with inflammatory and neuropathic pain. Previously, we described the identification of a series of amide-quinolinone iNOS dimerization inhibitors that although potent, suffered from high clearance and limited exposure in vivo. By conformationally restricting the amide of this progenitor series, we describe the identification of a novel series of benzimidazole-quinolinone dual iNOS/nNOS inhibitors with low clearance and sustained exposure in vivo. Compounds were triaged utilizing an LPS challenge assay coupled with mouse and rhesus pharmacokinetics and led to the identification of 4,7-imidazopyrazine 42 as the lead compound. 42 (KD7332) (J. Med. Chem. 2009, 52, 3047 - 3062) was confirmed as an iNOS dimerization inhibitor and was efficacious in the mouse formalin model of nociception and Chung model of neuropathic pain, without showing tolerance after repeat dosing. Further 42 did not affect motor coordination up to doses of 1000 mg/kg, demonstrating a wide therapeutic margin.


Assuntos
Analgésicos/síntese química , Fluoroquinolonas/síntese química , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Dor/tratamento farmacológico , Pirazinas/síntese química , Administração Oral , Analgésicos/química , Analgésicos/farmacologia , Animais , Linhagem Celular , Tolerância a Medicamentos , Fluoroquinolonas/química , Fluoroquinolonas/farmacologia , Humanos , Técnicas In Vitro , Camundongos , Microssomos Hepáticos/metabolismo , Dor/etiologia , Medição da Dor , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/etiologia , Multimerização Proteica , Pirazinas/química , Pirazinas/farmacologia , Teste de Desempenho do Rota-Rod , Relação Estrutura-Atividade
18.
J Med Chem ; 52(9): 3047-62, 2009 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19374401

RESUMO

There are three isoforms of dimeric nitric oxide synthases (NOS) that convert arginine to citrulline and nitric oxide. Inducible NOS is implicated in numerous inflammatory diseases and, more recently, in neuropathic pain states. The majority of existing NOS inhibitors are either based on the structure of arginine or are substrate competitive. We describe the identification from an ultra high-throughput screen of a novel series of quinolinone small molecule, nonarginine iNOS dimerization inhibitors. SAR studies on the screening hit, coupled with an in vivo lipopolysaccharide (LPS) challenge assay measuring plasma nitrates and drug levels, rapidly led to the identification of compounds 12 and 42--potent inhibitors of the human and mouse iNOS enzyme that were highly selective over endothelial NOS (eNOS). Following oral dosing, compounds 12 and 42 gave a statistical reduction in pain behaviors in the mouse formalin model, while 12 also statistically reduced neuropathic pain behaviors in the chronic constriction injury (Bennett) model.


Assuntos
Descoberta de Drogas , Fluoroquinolonas/administração & dosagem , Fluoroquinolonas/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Dor/tratamento farmacológico , Multimerização Proteica/efeitos dos fármacos , Pirazinas/administração & dosagem , Pirazinas/farmacologia , Quinolonas/administração & dosagem , Quinolonas/farmacologia , Administração Oral , Animais , Linhagem Celular , Constrição Patológica/induzido quimicamente , Constrição Patológica/tratamento farmacológico , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Fluoroquinolonas/química , Fluoroquinolonas/uso terapêutico , Formaldeído/toxicidade , Humanos , Concentração Inibidora 50 , Lipopolissacarídeos/toxicidade , Camundongos , Óxido Nítrico Sintase Tipo II/química , Óxido Nítrico Sintase Tipo II/metabolismo , Estrutura Quaternária de Proteína , Pirazinas/química , Pirazinas/uso terapêutico , Quinolonas/química , Quinolonas/uso terapêutico , Relação Estrutura-Atividade , Especificidade por Substrato
19.
Curr Chem Genomics ; 2: 1-9, 2008 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-20161838

RESUMO

The transcription of inducible nitric oxide synthase (iNOS) is activated by a network of proinflammatory signaling pathways. Here we describe the identification of a small molecule that downregulates the expression of iNOS mRNA and protein in cytokine-activated cells and suppresses nitric oxide production in vivo. Mechanistic analysis suggests that this small molecule, erstressin, also activates the unfolded protein response (UPR), a signaling pathway triggered by endoplasmic reticulum stress. Erstressin induces rapid phosphorylation of eIF2alpha and the alternative splicing of XBP-1, hallmark initiating events of the UPR. Further, erstressin activates the transcription of multiple genes involved in the UPR. These data suggest an inverse relationship between UPR activation and iNOS mRNA and protein expression under proinflammatory conditions.

20.
Genes Dev ; 17(8): 977-90, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12672694

RESUMO

In Caenorhabditis elegans, an X chromosome-counting mechanism specifies sexual fate. Specific genes termed X-signal elements, which are present on the X chromosome, act in a concerted dose-dependent fashion to regulate levels of the developmental switch gene xol-1. In turn, xol-1 levels determine sexual fate and the activation state of the dosage compensation mechanism. The crystal structure of the XOL-1 protein at 1.55 A resolution unexpectedly reveals that xol-1 encodes a GHMP kinase family member, despite sequence identity of 10% or less. Because GHMP kinases, thus far, have only been characterized as small molecule kinases involved in metabolic pathways, for example, amino acid and cholesterol synthesis, XOL-1 is the first member that controls nonmetabolic processes. Biochemical investigations demonstrated that XOL-1 does not bind ATP under standard conditions, suggesting that XOL-1 acts by a mechanism distinct from that of other GHMP kinases. In addition, we have cloned a XOL-1 ortholog from Caenorhabditis briggsae, a related nematode that diverged from C. elegans approximately 50-100 million years ago. These findings demonstrate an unanticipated role for GHMP kinase family members as mediators of sexual differentiation and dosage compensation and, possibly, other aspects of differentiation and development.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans/genética , Caenorhabditis/genética , Mecanismo Genético de Compensação de Dose , Proteínas de Helminto/química , Proteínas de Helminto/fisiologia , Processos de Determinação Sexual , Cromossomo X , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Caenorhabditis/crescimento & desenvolvimento , Caenorhabditis/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Clonagem Molecular , Cristalização , Cristalografia por Raios X , Transtornos do Desenvolvimento Sexual/genética , Dados de Sequência Molecular , Fosfotransferases (Aceptor do Grupo Álcool)/química , Fosfotransferases (Aceptor do Grupo Fosfato)/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Espectrometria de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA