Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Biochemistry ; 63(10): 1241-1245, 2024 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-38724483

RESUMEN

Natural products are important sources of seed compounds for drug discovery. However, it has become difficult in recent years to discover new compounds with valuable pharmacological activities. On the other hand, among the vast number of natural products that have been isolated so far, a considerable number of compounds with specific biological activities are thought to be overlooked in screening that uses biological activity as an index. Therefore, it is conceivable that such overlooked useful compounds may be found by screening compound libraries that have been amassed previously through specific assays. Previously, NPD723, a member of the Natural Products Depository library comprised of a mixture of natural and non-natural products developed at RIKEN, and its metabolite H-006 were found to inhibit growth of various cancer cells at low nanomolar half-maximal inhibitory concentration. Subsequent analysis revealed that H-006 strongly inhibited human dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme in the de novo pyrimidine biosynthetic pathway. Here, we elucidated the crystal structure of the DHODH-flavin mononucleotide-orotic acid-H-006 complex at 1.7 Å resolution to determine that furocoumavirin, the S-enantiomer of H-006, was the actual inhibitor. The overall mode of interaction of furocoumavirin with the inhibitor binding pocket was similar to that described for previously reported tight-binding inhibitors. However, the structural information together with kinetic characterizations of site-specific mutants identified key unique features that are considered to contribute to the sub-nanomolar inhibition of DHODH by furocoumavirin. Our finding identified new chemical features that could improve the design of human DHODH inhibitors.


Asunto(s)
Antivirales , Dihidroorotato Deshidrogenasa , Humanos , Antivirales/farmacología , Antivirales/química , Cristalografía por Rayos X , Dihidroorotato Deshidrogenasa/antagonistas & inhibidores , Dihidroorotato Deshidrogenasa/química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Furocumarinas/farmacología , Furocumarinas/química , Modelos Moleculares
2.
Oncol Res ; 31(6): 833-844, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37744270

RESUMEN

Dihydroorotate dehydrogenase (DHODH) is a central enzyme of the de novo pyrimidine biosynthesis pathway and is a promising drug target for the treatment of cancer and autoimmune diseases. This study presents the identification of a potent DHODH inhibitor by proteomic profiling. Cell-based screening revealed that NPD723, which is reduced to H-006 in cells, strongly induces myeloid differentiation and inhibits cell growth in HL-60 cells. H-006 also suppressed the growth of various cancer cells. Proteomic profiling of NPD723-treated cells in ChemProteoBase showed that NPD723 was clustered with DHODH inhibitors. H-006 potently inhibited human DHODH activity in vitro, whereas NPD723 was approximately 400 times less active than H-006. H-006-induced cell death was rescued by the addition of the DHODH product orotic acid. Moreover, metabolome analysis revealed that H-006 treatment promotes marked accumulation of the DHODH substrate dihydroorotic acid. These results suggest that NPD723 is reduced in cells to its active metabolite H-006, which then targets DHODH and suppresses cancer cell growth. Thus, H-006-related drugs represent a potentially powerful treatment for cancer and other diseases.


Asunto(s)
Dihidroorotato Deshidrogenasa , Proteómica , Humanos , Transformación Celular Neoplásica , Ciclo Celular , Muerte Celular
3.
ACS Chem Biol ; 18(2): 396-403, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36692171

RESUMEN

Proteasome inhibitors with γ-lactam structure, such as lactacystin and salinosporamide A, have been isolated from actinomycetes and have attracted attention as lead compounds for anticancer drugs. Previously, we identified a unique enzyme TAS1, which is the first reported fungal NRPS-PKS hybrid enzyme, from the filamentous fungus Pyricularia oryzae for the biosynthesis of a mycotoxin tenuazonic acid, a tetramic acid compound without γ-lactam structure. Homologues of TAS1 have been identified in several fungal genomes and classified into four groups (A-D). Here, we show that the group D TAS1 homologues from two filamentous fungi can biosynthesize γ-lactam compounds, taslactams A-D, with high similarity to actinomycete proteasome inhibitors. One of the γ-lactam compounds, taslactam C, showed potent proteasome inhibitory activity. In contrast to actinomycete γ-lactam compounds which require multiple enzymes for biosynthesis, the TAS1 homologue alone was sufficient for the biosynthesis of the fungal γ-lactam compounds.


Asunto(s)
Actinobacteria , Micotoxinas , Inhibidores de Proteasoma/farmacología , Lactamas/química , Péptido Sintasas/química
4.
ACS Chem Biol ; 17(2): 483-491, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35128925

RESUMEN

Glutathione peroxidase 4 (GPX4) is an intracellular enzyme that oxidizes glutathione while reducing lipid peroxides and is a promising target for cancer therapy. To date, several GPX4 inhibitors have been reported to exhibit cytotoxicity against cancer cells. However, some cancer cells are less sensitive to the known GPX4 inhibitors. This study aimed to explore compounds showing synergistic effects with GPX4 inhibitors. We screened a chemical library and identified a compound named NPD4928, whose cytotoxicity was enhanced in the presence of a GPX4 inhibitor. Furthermore, we identified ferroptosis suppressor protein 1 as its target protein. The results indicate that NPD4928 enhanced the sensitivity of various cancer cells to GPX4 inhibitors, suggesting that the combination might have therapeutic potential via the induction of ferroptosis.


Asunto(s)
Ferroptosis , Glutatión/metabolismo , Oxidación-Reducción , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Bibliotecas de Moléculas Pequeñas/farmacología
5.
ACS Chem Biol ; 16(11): 2570-2580, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34730931

RESUMEN

Dihydroorotate dehydrogenase (DHODH) catalyzes the rate-limiting step in de novo pyrimidine biosynthesis and is a promising cancer treatment target. This study reports the identification of indoluidin D and its derivatives as inhibitors of DHODH. Cell-based phenotypic screening revealed that indoluidin D promoted myeloid differentiation and inhibited the proliferation of acute promyelocytic leukemia HL-60 cells. Indoluidin D also suppressed cell growth in various other types of cancer cells. Cancer cell sensitivity profiling with JFCR39 and proteomic profiling with ChemProteoBase revealed that indoluidin D is a DHODH inhibitor. Indoluidin D inhibited human DHODH activity in vitro; the DHODH reaction product orotic acid rescued indoluidin D-induced cell differentiation. We synthesized several indoluidin D diastereomer derivatives and demonstrated that stereochemistry was vital to their molecular activity. The indoluidin D derivative indoluidin E showed similar activity to its parent compound and suppressed tumor growth in a murine lung cancer xenograft model. Hence, indoluidin D and its derivatives selectively inhibit DHODH and suppress cancer cell growth.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Dihidroorotato Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Animales , Antineoplásicos/química , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Bases de Datos de Proteínas , Inhibidores Enzimáticos/química , Humanos , Ratones , Proteómica , Estereoisomerismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
ACS Chem Biol ; 16(8): 1576-1586, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34296611

RESUMEN

Cancer cells reprogram their metabolism to survive and grow. Small-molecule inhibitors targeting cancer are useful for studying its metabolic pathways and functions and for developing anticancer drugs. Here, we discovered that glutipyran and its derivatives inhibit glycolytic activity and cell growth in human pancreatic cancer cells. According to proteomic profiling of glutipyran-treated cells using our ChemProteoBase, glutipyran was clustered within the group of endoplasmic reticulum (ER) stress inducers that included glycolysis inhibitors. Glutipyran inhibited glucose uptake and suppressed the growth of various cancer cells, including A431 cells that express glucose transporter class I (GLUT1) and DLD-1 GLUT1 knockout cells. When cotreated with the mitochondrial respiration inhibitor metformin, glutipyran exhibited a synergistic antiproliferative effect. Metabolome analysis revealed that glutipyran markedly decreased most metabolites of the glycolytic pathway and the pentose phosphate pathway. Glutipyran significantly suppressed tumor growth in a xenograft mouse model of pancreatic cancer. These results suggest that glutipyran acts as a broad-spectrum GLUT inhibitor and reduces cancer cell growth.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Facilitadoras del Transporte de la Glucosa/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Piranos/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Metabolómica , Metformina/uso terapéutico , Ratones Endogámicos BALB C , Ratones Desnudos , Proteómica , Piranos/síntesis química , Piranos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Am J Orthod Dentofacial Orthop ; 157(5): 680-689, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32354441

RESUMEN

INTRODUCTION: Osteoprotegerin-deficient mice develop severe high-turnover osteoporosis with porous low-density trabecular bone from an age-related increase in osteoclast activity and are useful alveolar bone models of osteoporosis or frail periodontal tissue. Bisphosphonate (BP), a first-line drug for osteoporosis, is bone-avid, causing side effects such as brittle and fragile bones and jaw osteonecrosis after tooth extraction. In orthodontics, active movement is precisely controlled by temporarily suppressing and resuming movement. BP impedes such control because of its long half-life of several years in bone. Therefore, we investigated the novel osteoclast-specific inhibitor reveromycin A (RMA), which has a short half-life in bone. We hypothesized that tooth movement could be precisely controlled through temporary discontinuation and re-administration of RMA. METHODS: Osteoprotegerin-deficient mice and wild-type mice were developed as tooth movement models under constant orthodontic force. A constant orthodontic force of 10 g was induced using a nickel-titanium closed coil spring to move the maxillary first molar for 14 days. We administered BP (1.25 mg/kg) or RMA (1.0 mg/kg) continuously and then discontinued it to reveal how the subsequent movement of teeth and surrounding alveolar bone was affected. RESULTS: Continuous BP or RMA administration suppressed osteoclast activity and preserved alveolar bone around the roots, apparently normalizing bone metabolism. Tooth movement remained suppressed after BP discontinuation but resumed at a higher rate after discontinuation of RMA. CONCLUSIONS: RMA appears useful for controlling orthodontic tooth movement because it can be suppressed and resumed through administration and discontinuation, respectively.


Asunto(s)
Compuestos de Espiro , Técnicas de Movimiento Dental , Animales , Remodelación Ósea , Ratones , Osteoclastos , Osteoprotegerina , Piranos
9.
Cell Chem Biol ; 27(2): 186-196.e4, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31813846

RESUMEN

The cellular thermal shift assay (CETSA) has recently been devised as a label-free method for target validation of small compounds and monitoring the thermal stabilization or destabilization of proteins due to binding with the compound. Herein, we developed a modified method by combining the CETSA and proteomics analysis based on 2D gel electrophoresis, namely 2DE-CETSA, to identify the thermal stability-shifted proteins by binding with a new compound. We applied the 2DE-CETSA for analysis of a target-unknown compound, NPD10084, which exerts anti-proliferative activity against colorectal cancer cells in vitro and in vivo, and identified pyruvate kinase muscle isoform 2 (PKM2) as a candidate target protein. Interestingly, NPD10084 interrupted protein-protein interactions between PKM2 and ß-catenin or STAT3, with subsequent suppression of downstream signaling. We thus demonstrate that our 2DE-CETSA method is applicable for identification of target compounds discovered by phenotypic screening.


Asunto(s)
Carbolinas/química , Proteínas Portadoras/metabolismo , Electroforesis en Gel Bidimensional/métodos , Proteínas de la Membrana/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Carbolinas/metabolismo , Carbolinas/farmacología , Carbolinas/uso terapéutico , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Unión Proteica/efectos de los fármacos , Proteoma/análisis , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Hormonas Tiroideas/genética , Trasplante Heterólogo , beta Catenina/química , beta Catenina/metabolismo , Proteínas de Unión a Hormona Tiroide
10.
J Antibiot (Tokyo) ; 72(12): 996-1000, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31481762

RESUMEN

Metarhizin C, a stereoisomer of BR-050 was isolated from a fungus Tolypocladium album RK17-F0007 through a screening program to search for new antitumor compounds. A structure of the isomer was determined by spectroscopic methods including detailed analysis of NOESY correlation and mass spectrometry, and found to be identical to that of 3-desacylmetarhizin A with the absolute structure. Previously, it had been isolated by Kikuchi et al and proposed as BR-050 including the stereo-structure. However, detailed analysis for the newly isolated isomer confirmed that 3-desacylmetarhizin A was not identical to BR-050. Therefore, we assigned it metarhizin C as a new BR-050 isomer. Metarhizin C showed selective cytotoxicity against osteosarcoma MG-63 cells in a glucose independent condition with IC50 value of 0.79 µg/ml, while > 30 µg/ml of IC50 value in a normal condition, and inhibited a mitochondrial respiration.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Hypocreales/metabolismo , Animales , Antimaláricos/química , Antimaláricos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Diterpenos/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Hypocreales/química , Hypocreales/aislamiento & purificación , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Ratas , Microbiología del Suelo , Estereoisomerismo
11.
Biochim Biophys Acta Proteins Proteom ; 1867(1): 28-37, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29883687

RESUMEN

Cancer cells can reprogram their metabolic machinery to survive. This altered metabolism, which is distinct from the metabolism of normal cells, is thought to be a possible target for the development of new cancer therapies. In this study, we constructed a screening system that focuses on bioenergetic profiles (specifically oxygen consumption rate and extracellular acidification rate) and characteristic proteomic changes. Thus, small molecules that target cancer-specific metabolism were investigated. We screened the chemical library of RIKEN Natural Products Depository (NPDepo) and found that unantimycin A, which was recently isolated from the fraction library of microbial metabolites, and NPL40330, which is derived from a chemical library, inhibit mitochondrial respiration. Furthermore, we developed an in vitro reconstitution assay method for mitochondrial electron transport chain using semi-intact cells with specific substrates for each complex of the mitochondrial electron transport chain. Our findings revealed that NPL40330 and unantimycin A target mitochondrial complexes I and III, respectively.


Asunto(s)
Descubrimiento de Drogas/métodos , Neoplasias/metabolismo , Proteómica/métodos , Animales , Descubrimiento de Drogas/tendencias , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/tendencias , Proteínas del Complejo de Cadena de Transporte de Electrón/efectos de los fármacos , Células HeLa , Humanos , Compuestos Macrocíclicos/farmacología , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fenotipo , Etiquetas de Fotoafinidad , Bibliotecas de Moléculas Pequeñas , Electroforesis Bidimensional Diferencial en Gel/métodos
12.
Sci Rep ; 7: 41710, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300073

RESUMEN

Osteoclasts degrade bone matrix proteins via the secretion of lysosomal enzymes. However, the precise mechanisms by which lysosomal components are transported and fused to the bone-apposed plasma membrane, termed ruffled border membrane, remain elusive. Here, we identified coronin 1A as a negative regulator of exocytotic release of cathepsin K, one of the most important bone-degrading enzymes in osteoclasts. The modulation of coronin 1A expression did not alter osteoclast differentiation and extracellular acidification, but strongly affected the secretion of cathepsin K and osteoclast bone-resorption activity, suggesting the coronin 1A-mediated regulation of lysosomal trafficking and protease exocytosis. Further analyses suggested that coronin 1A prevented the lipidation-mediated sorting of the autophagy-related protein LC3 to the ruffled border and attenuated lysosome-plasma membrane fusion. In this process, the interactions between coronin 1A and actin were crucial. Collectively, our findings indicate that coronin 1A is a pivotal component that regulates lysosomal fusion and the secretion pathway in osteoclast-lineage cells and may provide a novel therapeutic target for bone diseases.


Asunto(s)
Resorción Ósea/metabolismo , Catepsina K/metabolismo , Lisosomas/metabolismo , Proteínas de Microfilamentos/metabolismo , Osteoclastos/metabolismo , Actinas/metabolismo , Animales , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/genética , Resorción Ósea/patología , Diferenciación Celular/genética , Expresión Génica , Regulación de la Expresión Génica , Ratones , Osteoclastos/citología , Unión Proteica , Transporte de Proteínas , Ligando RANK/metabolismo
13.
Chem Biol Drug Des ; 89(6): 862-869, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27863017

RESUMEN

The human mutT homolog-1 (MTH1) protein prevents the incorporation of oxidized nucleotides such as 2-OH-dATP and 8-oxo-dGTP during DNA replication by hydrolyzing them into their corresponding monophosphates. It was found previously that cancer cells could tolerate oxidative stress due to this enzymatic activity of MTH1 and its inhibition could be a promising approach to treat several types of cancer. This finding has been challenged recently with increasing line of evidence suggesting that the cancer cell-killing effects of MTH1 inhibitors may be related to their engagement of off-targets. We have previously reported a few purine-based MTH1 inhibitors that enabled us to elucidate the dispensability of MTH1 in cancer cell survival. Here, we provide a detailed process of the identification of purine-based MTH1 inhibitors. Several new compounds with potency in the submicromolar range are disclosed. Furthermore, the structure-activity relationship and associated binding mode prediction using molecular docking have provided insights for the development of highly potent MTH1 inhibitors.


Asunto(s)
Enzimas Reparadoras del ADN/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Purinas/química , Purinas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad
14.
Sci Rep ; 6: 38385, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27922079

RESUMEN

Collismycin A (CMA), a microbial product, has anti-proliferative activity against cancer cells, but the mechanism of its action remains unknown. Here, we report the identification of the molecular target of CMA by ChemProteoBase, a proteome-based approach for drug target identification. ChemProteoBase profiling showed that CMA is closely clustered with di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone, an iron chelator. CMA bound to both Fe(II) and Fe(III) ions and formed a 2:1 chelator-iron complex with a redox-inactive center. CMA-induced cell growth inhibition was completely canceled by Fe(II) and Fe(III) ions, but not by other metal ions such as Zn(II) or Cu(II). Proteomic and transcriptomic analyses showed that CMA affects the glycolytic pathway due to the accumulation of HIF-1α. These results suggest that CMA acts as a specific iron chelator, leading to the inhibition of cancer cell growth.


Asunto(s)
2,2'-Dipiridil/análogos & derivados , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Quelantes del Hierro/farmacología , Hierro/química , Transcriptoma , 2,2'-Dipiridil/química , 2,2'-Dipiridil/aislamiento & purificación , 2,2'-Dipiridil/farmacología , Células A549 , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Bases de Datos de Compuestos Químicos , Glucólisis/efectos de los fármacos , Glucólisis/genética , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/agonistas , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hierro/metabolismo , Quelantes del Hierro/química , Quelantes del Hierro/aislamiento & purificación , Proteómica/métodos , Streptomyces/química , Tiosemicarbazonas/química , Tiosemicarbazonas/farmacología
15.
Sci Rep ; 6: 26521, 2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27210421

RESUMEN

Since recent publications suggested that the survival of cancer cells depends on MTH1 to avoid incorporation of oxidized nucleotides into the cellular DNA, MTH1 has attracted attention as a potential cancer therapeutic target. In this study, we identified new purine-based MTH1 inhibitors by chemical array screening. However, although the MTH1 inhibitors identified in this study targeted cellular MTH1, they exhibited only weak cytotoxicity against cancer cells compared to recently reported first-in-class inhibitors. We performed proteomic profiling to investigate the modes of action by which chemically distinct MTH1 inhibitors induce cancer cell death, and found mechanistic differences among the first-in-class MTH1 inhibitors. In particular, we identified tubulin as the primary target of TH287 and TH588 responsible for the antitumor effects despite the nanomolar MTH1-inhibitory activity in vitro. Furthermore, overexpression of MTH1 did not rescue cells from MTH1 inhibitor-induced cell death, and siRNA-mediated knockdown of MTH1 did not suppress cancer cell growth. Taken together, we conclude that the cytotoxicity of MTH1 inhibitors is attributable to off-target effects and that MTH1 is not essential for cancer cell survival.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Neoplasias/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteómica/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Supervivencia Celular/efectos de los fármacos , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/genética , Células HeLa , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/genética , Pirimidinas/farmacología , Tubulina (Proteína)/metabolismo
16.
FEBS Lett ; 590(8): 1152-62, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27001232

RESUMEN

We screened small-molecule compounds that inhibit osteoclast differentiation to find new anti-osteoporosis agents and found that a novel compound, SUKU-1, suppressed osteoclastogenesis. We also synthesized 38 derivatives of SUKU-1 and discovered that nine of them had inhibitory effects on osteoclastogenesis and that SUKU-33 was the most potent inhibitor. Next, we investigated the mechanisms by which SUKU-33 suppressed osteoclast differentiation. By measuring the uptake of [(3) H]-uridine in cells, we found that SUKU-33 suppressed both equilibrative nucleoside transporters and concentrative nucleoside transporters. These results suggest that SUKU-33 inhibits osteoclast differentiation by suppressing nucleoside transporters.


Asunto(s)
Proteínas de Transporte de Nucleósidos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos ICR , Osteoclastos/efectos de los fármacos , Osteogénesis/genética , Ligando RANK/farmacología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Tritio/metabolismo
17.
Sci Rep ; 5: 16510, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26561427

RESUMEN

Chronic periodontal disease is characterized by alveolar bone loss and inflammatory changes. Reveromycin A (RMA) was recently developed and is a unique agent for inhibiting osteoclast activity. This study analysed the effects of RMA in an experimental mouse model of periodontitis involving osteoprotegerin (OPG)-knockout mice, specifically, whether it could control osteoclasts and reduce inflammation in periodontal tissue. We examined wild-type (WT) and OPG knockout mice (OPG KO) ligated with wire around contact points on the left first and second molars. RMA was administered twice a day to half of the mice. Using micro-computed tomography, we measured the volume of alveolar bone loss between the first and second molars, and also performed histological analysis. The OPG KO RMA+ group had significantly decreased osteoclast counts, alveolar bone loss, attachment loss, and inflammatory cytokine expression 8 weeks after ligation. Thus, RMA may reduce alveolar bone loss and inflamed periodontal tissues in patients with periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar/etiología , Pérdida de Hueso Alveolar/patología , Osteoprotegerina/deficiencia , Enfermedades Periodontales/complicaciones , Enfermedades Periodontales/genética , Piranos/farmacología , Compuestos de Espiro/farmacología , Pérdida de Hueso Alveolar/diagnóstico , Pérdida de Hueso Alveolar/tratamiento farmacológico , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Noqueados , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Enfermedades Periodontales/diagnóstico , Enfermedades Periodontales/metabolismo , Microtomografía por Rayos X
18.
Nihon Rinsho ; 73(8): 1273-80, 2015 Aug.
Artículo en Japonés | MEDLINE | ID: mdl-26281678

RESUMEN

Target-based screening and cell-based screening are major approaches to identify anticancer drug candidates. Cell-based screening often contributes to the discovery of first-in-class drugs, but identification of the cellular targets of obtained compounds is a time-consuming step. To overcome this problem, affinity purification with small-molecule probes, which is a classic, but still the most common approach, has become more sophisticated and diversified. In addition, recent advances in omics studies and imaging analyses have allowed us to profile the biological effects of small molecules globally and quantitatively. Consequently, new therapeutic targets/drug leads involved in cancer cell cycle, transcription and redox regulation have been discovered.


Asunto(s)
Antineoplásicos , Descubrimiento de Drogas , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Aminoquinolinas/farmacología , Aminoquinolinas/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carbamatos/farmacología , Carbamatos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Cumarinas/farmacología , Cumarinas/uso terapéutico , Diaminas/farmacología , Diaminas/uso terapéutico , Diterpenos de Tipo Kaurano/farmacología , Diterpenos de Tipo Kaurano/uso terapéutico , Humanos , Neoplasias/patología , Pirazoles/farmacología , Pirazoles/uso terapéutico , Quinolinas/farmacología , Quinolinas/uso terapéutico
19.
Biosci Biotechnol Biochem ; 79(10): 1597-602, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25988721

RESUMEN

Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases that degrade many extracellular matrix components and that have been implicated in the pathogenesis of various human diseases including cancer metastasis. Here, we screened MMP-9 inhibitors using photo-cross-linked chemical arrays, which can detect small-molecule ligand-protein interactions on a chip in a high-throughput manner. The array slides were probed sequentially with His-MMP-9, anti-His antibody, and a Cy5-labeled secondary antibody and then scanned with a microarray scanner. We obtained 27 hits among 24,275 compounds from the NPDepo library; 2 of the identified compounds (isoxazole compound 1 and naphthofluorescein) inhibited MMP-9 enzyme activity in vitro. We further explored 17 analogs of 1 and found that compound 18 had the strongest inhibitory activity. Compound 18 also inhibited other MMPs, including MMP-2, MMP-12, and MMP-13 and significantly inhibited cell migration in human fibrosarcoma HT1080 cells. These results suggest that 18 is a broad-spectrum MMP inhibitor.


Asunto(s)
Fibroblastos/efectos de los fármacos , Fluoresceínas/farmacología , Isoxazoles/farmacología , Metaloproteinasa 9 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Descubrimiento de Drogas , Matriz Extracelular/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Fluoresceínas/química , Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Histidina/genética , Histidina/metabolismo , Humanos , Isoxazoles/química , Metaloproteinasa 12 de la Matriz/química , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 12 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/química , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/química , Análisis por Micromatrices , Oligopéptidos/genética , Oligopéptidos/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Bibliotecas de Moléculas Pequeñas/química
20.
J Med Chem ; 58(10): 4230-41, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25938266

RESUMEN

A series of indenopyrazoles was synthesized from the corresponding indanones and phenyl isothiocyanates in two steps. Among the compounds synthesized, methyl 3-((6-methoxy-1,4-dihydroindeno[1,2-c]pyrazol-3-yl)amino)benzoate 6m (GN39482) was found to possess a promising antiproliferative activity toward human cancer cells without affecting any antimicrobial and antimalarial activities at 100 nM. Both a methoxy group at R(1) position and a methoxycarbonyl group at R(2) position of the anilinoquinazoline framework are essential for the high cell growth inhibition. Both MorphoBase and ChemProteoBase profiling analyses suggested that compound 6m was classified as a tubulin inhibitor. Indeed, compound 6m inhibited the acetylated tubulin accumulation and the microtubule formation and induced G2/M cell cycle arrest in HeLa cells, revealing that a promising antiproliferative activity of compound 6m toward human cancer cells is probably caused by the tubulin polymerization inhibition.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Pirazoles/farmacología , Relación Estructura-Actividad , Moduladores de Tubulina/farmacología , metaminobenzoatos/farmacología , Antiinfecciosos/química , Antiinfecciosos/farmacología , Antimaláricos/química , Antimaláricos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Técnicas de Química Sintética , Células HeLa/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Plasmodium falciparum/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/química , Moduladores de Tubulina/química , metaminobenzoatos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA