Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 458: 116328, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36455640

RESUMEN

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS) that remains incurable. Herein, we demonstrated that ilepcimide (Antiepilepsirine), an antiepileptic drug used for decades, protects mice from experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Our studies found that ilepcimide treatment effectively ameliorates demyelination, blood-brain barrier leakage and infiltration of CD4+ and CD8+ T cells in EAE mice. On the one hand, ilepcimide can inhibit dihydroorotate dehydrogenase (DHODH), an important therapeutic target for MS. Computer molecular docking, thermal shift and fluorescence quenching assay demonstrated the directly interaction between ilepcimide and DHODH. Accordingly, ilepcimide observably repressed T cell proliferation in mixed lymphocyte reaction (MLR) assay and concanavalin A (Con-A) model in a DHODH-dependent manner. On the other hand, ilepcimide exhibited neuroprotective effect possibly through activating NRF2 antioxidant pathway in mouse neural crest-derived Neuro2a cells. Collectively, our findings have revealed the therapeutic potential of ilepcimide in EAE mouse model via restricting inflammatory response and oxidative stress, offering a potential opportunity for repurposing existing drug ilepcimide for MS therapy.


Asunto(s)
Reposicionamiento de Medicamentos , Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Ratones , Linfocitos T CD8-positivos , Dihidroorotato Deshidrogenasa , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Esclerosis Múltiple/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Inflamación/tratamiento farmacológico
2.
BMC Pulm Med ; 22(1): 324, 2022 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-36008805

RESUMEN

BACKGROUND: N6-methyladenosine (m6A) is the most common RNA modification, which plays a pivotal role in tumor development and progression. In this study, we assessed the role of m6A methyltransferase METTL3 in FRAS1-involved cell proliferation and colony formation of non-small cell lung cancer (NSCLC) cell lines. METHODS: Cell viability was analyzed by Cell Counting Kit (CCK-8) and colony formation. M6A RNA immunoprecipitation (IP), Ribosomal immunoprecipitation, RNA immunoprecipitation (RIP) were performed to verify the relationship between METTL3, FRAS1 and YTHDF1. Rescue experiments to confirm the regulatory mechanism of METTL3-FRAS1 promoted NSCLC cell proliferation through CDON by cooperating YTHDF1. RESULTS: We found that FRAS1 was correlated with poor prognosis in NSCLC patients, of which the transcript undergoes m6A modification regulated by METTL3. METTL3 silence reduced cell viability of NSCLC cells HCC827 and NCI-H1975, which could be restored by FRAS1 overexpression. The m6A modification of FRAS1 could be recognized by YTHDF1. FRAS1 silence or YTHDF1 silence could rescue the elevated NSCLC cell proliferation, colony formation, and tumor growth induced by METTL3 overexpression in vitro and in vivo. CONCLUSIONS: Our study reveals that METTL3-FRAS1 plays a crucial role in NSCLC cell proliferation, colony formation, and tumor growth through the regulation of CDON by cooperating YTHDF1.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Metiltransferasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metiltransferasas/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética
3.
Molecules ; 27(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35208999

RESUMEN

Metastatic pancreatic cancer remains a major clinical challenge, emphasizing the urgent need for the exploitation of novel therapeutic approaches with superior response. In this study, we demonstrate that the aberrant activation of prostaglandin E2 (PGE2) receptor 4 (EP4) is a pro-metastatic signal in pancreatic cancer. To explore the therapeutic role of EP4 signaling, we developed a potent and selective EP4 antagonist L001 with single-nanomolar activity using a panel of cell functional assays. EP4 antagonism by L001 effectively repressed PGE2-elicited cell migration and the invasion of pancreatic cancer cells in a dose-dependent manner. Importantly, L001 alone or combined with the chemotherapy drug gemcitabine exhibited remarkably anti-metastasis activity in a pancreatic cancer hepatic metastasis model with excellent tolerability and safety. Mechanistically, EP4 blockade by L001 abrogated Yes-associated protein 1 (YAP)-driven pro-metastatic factor expression in pancreatic cancer cells. The suppression of YAP's activity was also observed upon L001 treatment in vivo. Together, these findings support the notions that EP4-YAP signaling axis is a vital pro-metastatic pathway in pancreatic cancer and that EP4 inhibition with L001 may deliver a therapeutic benefit for patients with metastatic pancreatic cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/química , Animales , Antineoplásicos/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Dinoprostona/metabolismo , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vía de Señalización Hippo/efectos de los fármacos , Humanos , Ratones , Modelos Biológicos , Estructura Molecular , Metástasis de la Neoplasia , Neoplasias Pancreáticas/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
PLoS Comput Biol ; 16(2): e1007701, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32101536

RESUMEN

Tumor-specific genomic alterations allow systematic identification of genetic interactions that promote tumorigenesis and tumor vulnerabilities, offering novel strategies for development of targeted therapies for individual patients. We develop an Individualized Network-based Co-Mutation (INCM) methodology by inspecting over 2.5 million nonsynonymous somatic mutations derived from 6,789 tumor exomes across 14 cancer types from The Cancer Genome Atlas. Our INCM analysis reveals a higher genetic interaction burden on the significantly mutated genes, experimentally validated cancer genes, chromosome regulatory factors, and DNA damage repair genes, as compared to human pan-cancer essential genes identified by CRISPR-Cas9 screenings on 324 cancer cell lines. We find that genes involved in the cancer type-specific genetic subnetworks identified by INCM are significantly enriched in established cancer pathways, and the INCM-inferred putative genetic interactions are correlated with patient survival. By analyzing drug pharmacogenomics profiles from the Genomics of Drug Sensitivity in Cancer database, we show that the network-predicted putative genetic interactions (e.g., BRCA2-TP53) are significantly correlated with sensitivity/resistance of multiple therapeutic agents. We experimentally validated that afatinib has the strongest cytotoxic activity on BT474 (IC50 = 55.5 nM, BRCA2 and TP53 co-mutant) compared to MCF7 (IC50 = 7.7 µM, both BRCA2 and TP53 wild type) and MDA-MB-231 (IC50 = 7.9 µM, BRCA2 wild type but TP53 mutant). Finally, drug-target network analysis reveals several potential druggable genetic interactions by targeting tumor vulnerabilities. This study offers a powerful network-based methodology for identification of candidate therapeutic pathways that target tumor vulnerabilities and prioritization of potential pharmacogenomics biomarkers for development of personalized cancer medicine.


Asunto(s)
Biología Computacional/métodos , Redes Reguladoras de Genes , Neoplasias/genética , Antineoplásicos/uso terapéutico , Proteína BRCA2/genética , Biomarcadores de Tumor , Sistemas CRISPR-Cas , Carcinogénesis , Línea Celular Tumoral , Exoma , Pruebas Genéticas , Genómica , Humanos , Concentración 50 Inhibidora , Modelos Teóricos , Mutación , Neoplasias/tratamiento farmacológico , Farmacogenética , Medicina de Precisión , Mapeo de Interacción de Proteínas , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/genética
5.
Phytother Res ; 35(1): 463-476, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32779300

RESUMEN

Triple-negative breast cancer (TNBC) is heterogeneous disease with a poor prognosis. It is therefore important to explore novel therapeutic agents to improve the clinical efficacy for TNBC. The inosine 5'-monophosphate dehydrogenase 2 (IMPDH2) is a rate-limiting enzyme in the de novo synthesis of guanine nucleotides. It is always overexpressed in many types of tumors, including TNBC and regarded as a potential target for cancer therapy. Through screening a library of natural products, we identified shikonin, a natural bioactive component of Lithospermum erythrorhizon, is a novel and selective IMPDH2 inhibitor. Enzymatic analysis using Lineweaver-Burk plot indicates that shikonin is a competitive inhibitor of IMPDH2. The interaction between shikonin and IMDPH2 was further investigated by thermal shift assay, fluorescence quenching, and molecular docking simulation. Shikonin treatment effectively inhibits the growth of human TNBC cell line MDA-MB-231, and murine TNBC cell line, 4T1 in a dose-dependent manner, which is impaired by exogenous supplementation of guanosine, a salvage pathway of purine nucleotides. Most importantly, IMPDH2 knockdown significantly reduced cell proliferation and conferred resistance to shikonin in TNBC. Collectively, our findings showed the natural product shikonin as a selective inhibitor of IMPDH2 with anti-TNBC activity, impelling its further study in clinical trials.


Asunto(s)
Inhibidores Enzimáticos/farmacología , IMP Deshidrogenasa/antagonistas & inhibidores , Naftoquinonas/farmacología , Neoplasias de la Mama Triple Negativas/patología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Lithospermum/química , Ratones , Simulación del Acoplamiento Molecular , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
6.
Am J Hum Genet ; 100(1): 5-20, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-27939638

RESUMEN

The allosteric regulation triggering the protein's functional activity via conformational changes is an intrinsic function of protein under many physiological and pathological conditions, including cancer. Identification of the biological effects of specific somatic variants on allosteric proteins and the phenotypes that they alter during tumor initiation and progression is a central challenge for cancer genomes in the post-genomic era. Here, we mapped more than 47,000 somatic missense mutations observed in approximately 7,000 tumor-normal matched samples across 33 cancer types into protein allosteric sites to prioritize the mutated allosteric proteins and we tested our prediction in cancer cell lines. We found that the deleterious mutations identified in cancer genomes were more significantly enriched at protein allosteric sites than tolerated mutations, suggesting a critical role for protein allosteric variants in cancer. Next, we developed a statistical approach, namely AlloDriver, and further identified 15 potential mutated allosteric proteins during pan-cancer and individual cancer-type analyses. More importantly, we experimentally confirmed that p.Pro360Ala on PDE10A played a potential oncogenic role in mediating tumorigenesis in non-small cell lung cancer (NSCLC). In summary, these findings shed light on the role of allosteric regulation during tumorigenesis and provide a useful tool for the timely development of targeted cancer therapies.


Asunto(s)
Regulación Alostérica/genética , Sitio Alostérico/genética , Genoma Humano/genética , Mutación Missense/genética , Neoplasias/genética , Proteoma/genética , Proteómica , Carcinogénesis/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias
7.
Phytother Res ; 34(10): 2766-2777, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32430958

RESUMEN

Natural product corynoline is a unique isoquinoline alkaloid extracted from traditional Chinese medicine Corydalis bungeana Turcz, whereas its anticancer properties have not been investigated. In this study, we found that corynoline potently impairs the growth of melanoma cells, B16F10, and A375 in a concentration-dependent manner. Treatment of melanoma cells with corynoline results in G2 cell arrest accompanied by reduced cdc2 activation. Furthermore, corynoline triggers apoptosis of melanoma cells, which is associated with increased expression of Bax and cleaved caspase-3. Mechanistic study indicates that corynoline strongly induces reactive oxygen species (ROS) generation and subsequent DNA damage as evidenced by γ-H2AX accumulation. Notably, the effect of corynoline on melanoma cell cycle and apoptosis is abolished by a ROS scavenger N-acetyl cysteine (NAC), indicating a ROS-dependent mechanism. Finally, corynoline significantly inhibits in vivo B16F10 melanoma tumor growth accompanied by reduced expression of Ki-67 in tumor tissue. Taken together, our data suggest that corynoline suppresses melanoma cell growth in vitro and in vivo by inducing oxidative stress and represents a potential therapeutic agent for melanoma patients.


Asunto(s)
Alcaloides de Berberina/uso terapéutico , Productos Biológicos/química , Medicina Tradicional China/métodos , Melanoma/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Alcaloides de Berberina/farmacología , Humanos
8.
J Orthop Sci ; 25(4): 708-714, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31607516

RESUMEN

BACKGROUND: The use of 3D-printed scaffolds in repairing bone defects remains unexplored. We aimed to determine whether the duration of electrochemical deposition (ECD) affects the properties of hydroxyapatite (HA) coatings on 3D-printed titanium (TI) scaffolds as well as the corresponding phenotype of MC3T3-E1 cells seeded on these surfaces. METHODS: Five groups of HA-coated TI scaffolds were produced using different durations of ECD (0, 5, 10, 20, and 30 min) and examined under scanning electron microscopy (SEM). MC3T3-E1 cell adhesion to the HA-coated scaffolds and subsequent proliferation and viability were assessed using SEM, DAPI staining, EdU staining, and Alamar Blue assay, respectively. MC3T3-E1 cell expression of osteogenic genes was analyzed by fluorescence RT-PCR. RESULTS: On SEM, longer ECD durations resulted in more compact HA crystals of differing morphology coated onto the TI scaffolds. MC3T3-E1 cell adhesion differed among the five groups (p < 0.05), with the largest number of cells adhered to the scaffolds prepared with 30 min of ECD, followed by the group prepared with 20 min of ECD. However, the ECD duration of 20 min was associated with the highest cell viability and proliferation rate (both p < 0.05) as well as the highest mRNA expression levels of alkaline phosphatase, collagen I, osteocalcin and runt-related transcription factor 2 among the five groups (p < 0.05). CONCLUSIONS: In the fabrication of HA-coated 3D printed TI scaffolds, an ECD duration of 20 min resulted in scaffolds that best promoted MC3T3-E1 cell viability, proliferation and osteogenic gene expression.


Asunto(s)
Materiales Biocompatibles , Adhesión Celular/fisiología , Durapatita , Osteogénesis , Impresión Tridimensional , Andamios del Tejido , Titanio , Células 3T3 , Animales , Ratones , Factores de Tiempo
9.
Haematologica ; 103(9): 1472-1483, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29880605

RESUMEN

Acute myeloid leukemia is a disorder characterized by abnormal differentiation of myeloid cells and a clonal proliferation derived from primitive hematopoietic stem cells. Interventions that overcome myeloid differentiation have been shown to be a promising therapeutic strategy for acute myeloid leukemia. In this study, we demonstrate that CRISPR/Cas9-mediated knockout of dihydroorotate dehydrogenase leads to apoptosis and normal differentiation of acute myeloid leukemia cells, indicating that dihydroorotate dehydrogenase is a potential differentiation regulator and a therapeutic target in acute myeloid leukemia. By screening a library of natural products, we identified a novel dihydroorotate dehydrogenase inhibitor, isobavachalcone, derived from the traditional Chinese medicine Psoralea corylifolia Using enzymatic analysis, thermal shift assay, pull down, nuclear magnetic resonance, and isothermal titration calorimetry experiments, we demonstrate that isobavachalcone inhibits human dihydroorotate dehydrogenase directly, and triggers apoptosis and differentiation of acute myeloid leukemia cells. Oral administration of isobavachalcone suppresses subcutaneous HL60 xenograft tumor growth without obvious toxicity. Importantly, our results suggest that a combination of isobavachalcone and adriamycin prolonged survival in an intravenous HL60 leukemia model. In summary, this study demonstrates that isobavachalcone triggers apoptosis and differentiation of acute myeloid leukemia cells via pharmacological inhibition of human dihydroorotate dehydrogenase, offering a potential therapeutic strategy for acute myeloid leukemia.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Animales , Apoptosis/genética , Biomarcadores de Tumor , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Chalconas/química , Chalconas/farmacología , Dihidroorotato Deshidrogenasa , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Ratones , Modelos Moleculares , Estructura Molecular , Células Madre Neoplásicas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Pronóstico , Interferencia de ARN , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Pharmacol Res ; 129: 400-413, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29133212

RESUMEN

G protein-coupled receptors (GPCRs) are the largest super family with more than 800 membrane receptors. Currently, over 30% of the approved drugs target human GPCRs. However, only approximately 30 human GPCRs have been resolved three-dimensional crystal structures, which limits traditional structure-based drug discovery. Recent advances in network-based systems pharmacology approaches have demonstrated powerful strategies for identifying new targets of GPCR ligands. In this study, we proposed a network-based systems pharmacology framework for comprehensive identification of new drug-target interactions on GPCRs. Specifically, we reconstructed both global and local drug-target interaction networks for human GPCRs. Network analysis on the known drug-target networks showed rational strategies for designing new GPCR ligands and evaluating side effects of the approved GPCR drugs. We further built global and local network-based models for predicting new targets of the known GPCR ligands. The area under the receiver operating characteristic curve of more than 0.96 was obtained for the best network-based models in cross validation. In case studies, we identified that several network-predicted GPCR off-targets (e.g. ADRA2A, ADRA2C and CHRM2) were associated with cardiovascular complications (e.g. bradycardia and palpitations) of the approved GPCR drugs via an integrative analysis of drug-target and off-target-adverse drug event networks. Importantly, we experimentally validated that two newly predicted compounds, AM966 and Ki16425, showed high binding affinities on prostaglandin E2 receptor EP4 subtype with IC50=2.67µM and 6.34µM, respectively. In summary, this study offers powerful network-based tools for identifying polypharmacology of GPCR ligands in drug discovery and development.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Simulación por Computador , Descubrimiento de Drogas , Humanos , Ligandos , Polifarmacología
12.
Wei Sheng Wu Xue Bao ; 57(3): 411-21, 2017 Mar 04.
Artículo en Zh | MEDLINE | ID: mdl-29756439

RESUMEN

Objective: Endophytes are widespread in plants and build long-term mutually beneficial symbiotic relationship with the host. However, the mechanism of their interactions with the host needs further study. To explore the mechanism of endophytic bacterium ginkgo endophyte KM-1-2, we managed to forecast its secretory proteins based on its genome and explicit characteristics. Methods: Signal peptide analysis software SignalP, transmembrane helical structure analysis software TMHMM and Phobius, cells position software PSORT, subcellar localization software TargetP and GPI anchor site analysis software big-PI Predictor were used to predict the scope of all secreted proteins, which were defined as secretome. Results: Altogether 128 typical signal peptide secretory proteins were screened out of 5299 protein sequences in KM-1-2 genome, accounting for 2.4% of the whole genome. The shortest ORF encoding these proteins is 61 bp, the longest one is 2105 bp and the average is 373 bp. The length of the signal peptide guiding secretory protein was distributed between 15 to 37 aa, with the average length of 24 aa. Amino acid with the highest present frequency of signal peptide in proper order is alanine, leucine and valine. The type of signal peptide cleavage belongs to A-X-A which named SPI cleavage type. Among the total secretory proteins 66 pieces have functional description and 26 pieces were enzymes. These enzymes mainly include glycoside hydrolase, esterase transferase, REDOX enzyme and carbon oxygen lyase. Conclusion: The predicted secretory proteins of Streptomyces lavendulae KM-1-2 were achieved through bioinformatics analysis. These secretory proteins involved some enzymes and other unknown functions. This result laid the foundation for further study between endophyte and host.


Asunto(s)
Proteínas Bacterianas/genética , Endófitos/genética , Ginkgo biloba/microbiología , Señales de Clasificación de Proteína , Streptomyces/genética , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/genética , Sistemas de Secreción Bacterianos/metabolismo , Endófitos/clasificación , Endófitos/aislamiento & purificación , Endófitos/metabolismo , Sistemas de Lectura Abierta , Transporte de Proteínas , Streptomyces/clasificación , Streptomyces/aislamiento & purificación , Streptomyces/metabolismo
13.
Biochem Biophys Res Commun ; 477(4): 915-922, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27378425

RESUMEN

Human inosine 5'-monophosphate dehydrogenase (hIMPDH) is a rate-limiting enzyme in the de novo biosynthetic pathway of purine nucleotides, playing crucial roles in cellular proliferation, differentiation, and transformation. Dysregulation of hIMPDH expression and activity have been found in a variety of human cancers including leukemia. In this study, we found that myricetin, a naturally occurring phytochemical existed in berries, wine and tea, was a novel inhibitor of human type 1 and type 2 IMPDH (hIMPDH1/2) with IC50 values of 6.98 ± 0.22 µM and 4.10 ± 0.14 µM, respectively. Enzyme kinetic analysis using Lineweaver-Burk plot revealed that myricetin is a mix-type inhibitor for hIMPDH1/2. Differential scanning fluorimetry and molecular docking simulation data demonstrate that myricetin is capable of binding with hIMPDH1/2. Myricetin treatment exerts potent anti-proliferative and pro-apoptotic effects on K562 human leukemia cells in a dose-dependent manner. Importantly, cytotoxicity of myricetin on K562 cells were markedly attenuated by exogenous addition of guanosine, a salvage pathway of maintaining intracellular pool of guanine nucleotides. Taking together, these results indicate that natural product myricetin exhibits potent anti-leukemia activity by interfering with purine nucleotides biosynthetic pathway through the suppression of hIMPDH1/2 catalytic activity.


Asunto(s)
Flavonoides/administración & dosificación , Flavonoides/química , IMP Deshidrogenasa/antagonistas & inhibidores , IMP Deshidrogenasa/metabolismo , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Sitios de Unión , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células K562 , Simulación del Acoplamiento Molecular , Terapia Molecular Dirigida/métodos , Unión Proteica , Resultado del Tratamiento
14.
J Pharmacol Exp Ther ; 357(3): 466-75, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27048660

RESUMEN

UNLABELLED: ß-Lapachone [ß-lap; 3,4-dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione] is a novel anticancer drug currently under investigation in phase I/II clinical trials. However, the mechanism underlying its clinical efficacy remains unclear. In this study, we found that ß-lap provoked the cleavage of heat shock protein 90 (Hsp90) in NAD(P)H: quinone oxidoreductase-1 (NQO1)-expressing lung and prostate cancer cells as well as in primary human umbilical vein endothelial cells (HUVECs). These actions of ß-lap were different from that of the conventional Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin. As a consequence of Hsp90 cleavage, Hsp90-associated oncoproteins, such as receptor-interacting protein, Raf-1, AKT, and CDK4, were degraded in treated cancer cells, and key receptor tyrosine kinases such as vascular endothelial cell growth factor receptor-2 and Her-2 were degraded in treated HUVECs through a proteasomal system. Further results revealed that specific inhibitors of NQO1 and reactive oxygen species could dramatically reduce ß-lap-mediated Hsp90 cleavage. In addition to its cytotoxicity, ß-lap effectively inhibited angiogenesis by suppressing tube formation and the invasion of HUVECs in vitro, rat aortic microvascular sprouts ex vivo, and mouse corneal neovascularization in vivo. Furthermore, ß-lap markedly suppressed the growth and angiogenesis of human lung cancer xenografts in nude mice and decreased the levels of receptor-interacting protein, AKT, CDK4, and CD31 in the solid tumors. Unlike other NQO1-dependent cytotoxic quinones, such as streptonigrin, menadione, mitomycin, and 17-allylamino-17-demethoxygeldanamycin, ß-lap was the only agent that could cause Hsp90 cleavage. Taken together, our results suggest a crucial mechanism underlying the antitumor efficacy of ß-lap.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/farmacología , Estrés Oxidativo/efectos de los fármacos , Proteolisis/efectos de los fármacos , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Aorta/efectos de los fármacos , Aorta/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Naftoquinonas/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Proteínas Oncogénicas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica/efectos de los fármacos , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Enzyme Inhib Med Chem ; 31(sup4): 98-105, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27590167

RESUMEN

Human lipoxygenases (LOXs) have been emerging as effective therapeutic targets for inflammatory diseases. In this study, we found that four natural 2-arylbenzo[b]furan derivatives isolated from Artocarpus heterophyllus exhibited potent inhibitory activities against human LOXs, including moracin C (1), artoindonesianin B-1 (2), moracin D (3), moracin M (4). In our in vitro experiments, compound 1 was identified as the most potent LOX inhibitor and the moderate subtype selective inhibitor of 12-LOX. Compounds 1 and 2 act as competitive inhibitors of LOXs. Moreover, 1 significantly inhibits LTB4 production and chemotactic capacity of neutrophils, and is capable of protecting vascular barrier from plasma leakage in vivo. In addition, the preliminary structure-activity relationship analysis was performed based on the above four naturally occurring (1-4) and six additional synthetic 2-arylbenzo[b]furan derivatives. Taken together, these 2-arylbenzo[b]furan derivatives, as LOXs inhibitors, could represent valuable leads for the future development of therapeutic agents for inflammatory diseases.


Asunto(s)
Benzofuranos/farmacología , Inhibidores de la Lipooxigenasa/química , Inhibidores de la Lipooxigenasa/farmacología , Lipooxigenasa/metabolismo , Artocarpus/química , Benzofuranos/química , Benzofuranos/aislamiento & purificación , Relación Dosis-Respuesta a Droga , Humanos , Lipooxigenasa/aislamiento & purificación , Inhibidores de la Lipooxigenasa/aislamiento & purificación , Estructura Molecular , Relación Estructura-Actividad
16.
Int J Mol Sci ; 17(8)2016 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-27463712

RESUMEN

Artocarpus heterophyllus, a popular tropical fruit commonly known as the jackfruit tree, is normally planted in subtropical or tropical areas. Since a variety of phytochemicals isolated from A. heterophyllus have been found to possess potently anti-inflammatory, antiviral and antimalarial activities, researchers have devoted much interest to its potential pharmaceutical value. However, the exact mechanism underlying its anti-inflammatory activity is not well characterized. In this study, seven natural products isolated from A. heterophyllus, including 25-Hydroxycycloart-23-en-3-one (HY), Artocarpin (AR), Dadahol A (DA), Morachalcone A (MA), Artoheterophyllin B (AB), Cycloheterophyllin (CY) and Moracin C (MC) were collected. Lipopolysaccharide (LPS)-stimulated inflammatory response in RAW264.7 macrophages were used in this study. Among these compounds, MC significantly inhibited LPS-activated reactive oxygen species (ROS) and nitric oxide (NO) release without marked cytotoxicity. Furthermore, MC effectively reduced LPS stimulated up-regulation of mRNA and protein expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and serval pro-inflammatory cytokines (interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumor necrosis factor α (TNF-α)). Mechanistic studies revealed that the anti-inflammatory effect of MC was associated with the activation of the mitogen activated protein kinases (MAPKs) (including p38, ERK and JNK) and nuclear factor-κB (NF-κB) pathways, especially reducing the nuclear translocation of NF-κB p65 subunit as revealed by nuclear separation experiment and confocal microscopy.


Asunto(s)
Antiinflamatorios/farmacología , Artocarpus/química , Benzofuranos/farmacología , Inflamación/inmunología , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Estilbenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/patología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Pharmacol Exp Ther ; 352(1): 129-38, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25352499

RESUMEN

Repurposing existing drugs not only accelerates drug discovery but rapidly advances clinical therapeutic strategies. In this article, we identified potassium antimonyl tartrate (PAT), an antiparasitic drug, as a novel agent to block angiogenesis by screening US Food and Drug Administration-approved chemical drugs. By comparing the cytotoxicity of PAT in various nonsmall-cell lung cancer (NSCLC) cells with that observed in primary cultured human umbilical vein endothelial cells (HUVECs), we found that HUVECs were much more sensitive to the PAT treatment. In in vivo tumor xenograft mouse models established either by PAT-resistant A549 cells or by patient primary tumors, PAT significantly decreased the tumor volume and tumor weight of NSCLC xenografts at dosage of 40 mg/kg (i.p., daily) and, more importantly, augmented the antitumor efficacy of cisplatin chemotherapy. Remarkable loss of vascularization in the treated xenografts indicated the in vivo antiangiogenesis property of PAT, which was well correlated with its tumor growth inhibition in NSCLC cells. Furthermore, in the in vitro angiogenic assays, PAT exhibited dose-dependent inhibition of HUVEC proliferation, migration, and tube formation in response to different stimuli. Consistently, PAT also abolished the vascular endothelial cell growth factor-induced angiogenesis in the Matrigel plugs assay. Mechanistically, we found that PAT inhibited the activities of several receptor tyrosine kinases and specifically blocked the activation of downstream Src and focal adhesion kinases in HUVECs. Taken together, our results characterized the novel antiangiogenic and antitumor function of PAT in NSCLC cells. Further study of PAT in anticancer clinical trials may be warranted.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Tartrato de Antimonio y Potasio/farmacología , Antiparasitarios/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Tartrato de Antimonio y Potasio/uso terapéutico , Antiparasitarios/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Cisplatino/farmacología , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Ratones , Proteínas Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Breast Cancer Res Treat ; 148(2): 279-89, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25301089

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is implicated breast cancer metastasis and represents a potential target for developing new anti-tumor metastasis drugs. The purpose of this study is to investigate whether the natural agent 1'-acetoxychavicol acetate (ACA), derived from the rhizomes and seeds of Languas galanga, could suppress breast cancer metastasis by targeting STAT3 signaling pathway. ACA was examined for its effects on breast cancer migration/invasion and metastasis using Transwell assays in vitro and breast cancer skeletal metastasis mouse model in vivo (n = 10 mice per group). The inhibitory effect of ACA on cellular STAT3 signaling pathway was investigated by series of biochemistry analysis. The chavicol preferentially suppressed cancer cell migration and invasion, and this activity was superior to its cytotoxic effects. ACA suppressed both constitutive and interleukin-6-inducible STAT3 activation and diminished the accumulation of STAT3 in the nucleus and its DNA-binding activity. More importantly, ACA treatment led to significant up-regulation of Src homology region 2 domain-containing phosphatase 1 (SHP-1), and the ACA-induced depression of cancer cell migration and STAT3 signaling could be apparently reversed by blockade of SHP-1. Matrix metalloproteinase (MMP)-2 and -9, gene products of STAT3 that regulate cell invasion, were specifically suppressed by ACA. In tumor metastasis model, ACA potently inhibited the human breast cancer cell-induced osteolysis, and had little apparent in vivo toxicity at the test concentrations. ACA is a novel drug candidate for the inhibition of tumor metastasis through interference with the SHP-1/STAT3/MMPs signaling pathway.


Asunto(s)
Alcoholes Bencílicos/uso terapéutico , Neoplasias Óseas/prevención & control , Neoplasias de la Mama/prevención & control , Metaloproteinasa 3 de la Matriz/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Metaloproteinasa 3 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Langmuir ; 30(34): 10248-55, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25105821

RESUMEN

Cross-linked polymer particles were prepared via surfactant-free emulsion copolymerization of 2-(diethylamino)ethyl methacrylate (DEAEMA) and sodium methacrylate (SMA) using N,N'-methylenebis(acrylamide) (MBA) as a cross-linker. Generated particles are zwitterionic, possessing unique isoelectric points in the pH range of 7.5-8.0, which is readily tunable through CO2/N2 bubbling. The particles were found to be highly responsive to CO2/N2 switching, dissolving in water with CO2 bubbling and precipitating with N2 bubbling at room temperature. Pickering emulsions of n-dodecane were prepared using these particles as the sole emulsifier. These emulsions can be rapidly demulsified with CO2 bubbling, resulting in complete oil/water phase separations. Nitrogen bubbling efficiently re-emulsifies the oil with the aid of homogenization. The rapid emulsification/demulsification using CO2/N2 bubbling at room temperature provides these cross-linked zwitterionic particles with distinct advantages as functional Pickering surfactants.


Asunto(s)
Dióxido de Carbono/química , Emulsiones , Nitrógeno/química , Temperatura , Microscopía Electrónica de Transmisión , Espectroscopía de Protones por Resonancia Magnética
20.
Org Biomol Chem ; 12(38): 7591-7, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25139438
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA