Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 15(7): e0234103, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32645016

RESUMEN

Cyclin-dependent kinases (CDKs) contribute to the cancer hallmarks of uncontrolled proliferation and increased survival. As a result, over the last two decades substantial efforts have been directed towards identification and development of pharmaceutical CDK inhibitors. Insights into the biological consequences of CDK inhibition in specific tumor types have led to the successful development of CDK4/6 inhibitors as treatments for certain types of breast cancer. More recently, a new generation of pharmaceutical inhibitors of CDK enzymes that regulate the transcription of key oncogenic and pro-survival proteins, including CDK9, have entered clinical development. Here, we provide the first disclosure of the chemical structure of fadraciclib (CYC065), a CDK inhibitor and clinical candidate designed by further optimization from the aminopurine scaffold of seliciclib. We describe its synthesis and mechanistic characterization. Fadraciclib exhibits improved potency and selectivity for CDK2 and CDK9 compared to seliciclib, and also displays high selectivity across the kinome. We show that the mechanism of action of fadraciclib is consistent with potent inhibition of CDK9-mediated transcription, decreasing levels of RNA polymerase II C-terminal domain serine 2 phosphorylation, the pro-survival protein Myeloid Cell Leukemia 1 (MCL1) and MYC oncoprotein, and inducing rapid apoptosis in cancer cells. This cellular potency and mechanism of action translate to promising anti-cancer activity in human leukemia mouse xenograft models. Studies of leukemia cell line sensitivity identify mixed lineage leukemia (MLL) gene status and the level of B-cell lymphoma 2 (BCL2) family proteins as potential markers for selection of patients with greater sensitivity to fadraciclib. We show that the combination of fadraciclib with BCL2 inhibitors, including venetoclax, is synergistic in leukemic cell models, as predicted from simultaneous inhibition of MCL1 and BCL2 pro-survival pathways. Fadraciclib preclinical pharmacology data support its therapeutic potential in CDK9- or CDK2-dependent cancers and as a rational combination with BCL2 inhibitors in hematological malignancies. Fadraciclib is currently in Phase 1 clinical studies in patients with advanced solid tumors (NCT02552953) and also in combination with venetoclax in patients with relapsed or refractory chronic lymphocytic leukemia (CLL) (NCT03739554) and relapsed refractory acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) (NCT04017546).


Asunto(s)
Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacología , Animales , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 9 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 9 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Humanos , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Sulfonamidas/farmacología
2.
Biochim Biophys Acta Gen Subj ; 1864(3): 129503, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31816347

RESUMEN

BACKGROUND: In recent times, coordination complexes of iron in various oxidation states along with variety of ligand systems have been designed and developed for effective treatment of cancer cells without adversely affecting the normal cell and tissues of various organs. METHODS: In this study, we have evaluated the mechanism of action of a Fe(II) Schiff base complex in the crop plant Trigonella foenum-graecum L. (Fenugreek) as the screening system by using morphological, cytological, biochemical and molecular approaches. Further functional characterization was performed using MCF-7 cell line and solid tumour model for the assessment of anti-tumour activity of the complex. RESULTS: Our results indicate efficiency of the Fe(II) Schiff base complex in the induction of double strand breaks in DNA. Complex treatment clearly induced cytotoxic and genotoxic damage in Trigonella seedlings. The Fe-complex treatment caused cell cycle arrest via the activation of ATM-ATR kinase mediated DNA damage response pathway with the compromised expression of CDK1, CDK2 and CyclinB1 protein in Trigonella seedlings. In cultured MCF-7 cells, the complex induces cytotoxicity and DNA fragmentation through intracellular ROS generation. Fe-complex treatment inhibited tumour growth in solid tumour model with no additional side effects. CONCLUSION: The growth inhibitory and cytotoxic effects of the complex result from activation of DNA damage response along with oxidative stress and cell cycle arrest. GENERAL SIGNIFICANCE: Overall, our results have provided comprehensive information on the mechanism of action and efficacy of a Fe(II) Schiff base complex in higher eukaryotic genomes and indicated its future implications as potential therapeutic agent.


Asunto(s)
Hierro/metabolismo , Trigonella/metabolismo , Proteína Quinasa CDC2/efectos de los fármacos , Ciclina B1/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Compuestos Ferrosos/metabolismo , Humanos , Células MCF-7/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Bases de Schiff/metabolismo , Trigonella/química
3.
Antiviral Res ; 172: 104621, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31634495

RESUMEN

HSV-1 is one of the most prevalent viruses worldwide, and due to the limited therapies mainly with acyclovir and analogues and the emergence of acyclovir (ACV) resistant strains, the search for new drugs with different modes of action is needed. This study identified compounds that bind in silico to cyclin dependent kinase 2 (CDK2), a cellular enzyme required for efficient HSV-1 replication, and have anti-HSV-1 activity. Compounds obtained from virtual screening by Pharmit were filtered in FAF-Drugs4 for good pharmacokinetic and toxicological profiles and submitted to molecular docking on CDK2 using Autodock Vina. The six most promising compounds were evaluated for inhibiting lytic replication of HSV-1 wild-type and ACV-resistant strains on human fibroblasts. The compounds were also assayed for cytotoxicity. Compounds 1, 2 and 3 showed antiviral activity with EC50 (50% of effective drug concentration) of 32, 29 and 64 µM and CC50 (50% of cytotoxic concentration) of 159, 1410 and 2044 µM, respectively. Compounds 1 and 2 were also active against ACV resistant strains and compound 3 inhibited the reactivation of HSV-1 in neurons, which is an important finding to guide drug design of new anti-HSV-1 antivirals with different modes of action. These compounds are promising candidates for optimization into more potent agents to treat HSV-1 infections and recurrences.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Herpesvirus Humano 1/efectos de los fármacos , Neuronas/virología , Inhibidores de Proteínas Quinasas , Antivirales/farmacocinética , Antivirales/toxicidad , Línea Celular , Diseño de Fármacos , Farmacorresistencia Viral/efectos de los fármacos , Herpes Simple/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/toxicidad , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
4.
Medicine (Baltimore) ; 97(4): e9653, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29369181

RESUMEN

Dexamethasone (DEX) could induce low birth weight of infant, and low birth weight has close associations with glucocorticoid levels, insulin resistance, hypertension, and metabolic syndrome in adulthood. This study was designed to reveal the action mechanisms of DEX on the birth weight of infant.Using quantitative real-time polymerase chain reaction (qRT-PCR), trophoblast cells of human placenta were identified and the optimum treatment time of DEX were determined. Trophoblast cells were treated by DEX (DEX group) or ethanol (control group) (each group had 3 samples), and then were performed with RNA-sequencing. Afterward, the differentially expressed genes (DEGs) were identified by R package, and their potential functions were successively enriched using DAVID database and Enrichr method. Followed by protein-protein interaction (PPI) network was constructed using Cytoscape software. Using Enrichr method and TargetScan software, the transcription factors (TFs) and micorRNAs (miRNAs) targeted the DEGs separately were predicted. Based on MsigDB database, gene set enrichment analysis (GSEA) was performed.There were 391 DEGs screened from the DEX group. Upregulated SRR and potassium voltage-gated channel subfamily J member 4 (KCNJ4) and downregulated GALNT1 separately were enriched in PDZ (an acronym of PSD-95, Dlg, and ZO-1) domain binding and Mucin type O-glycan biosynthesis. In the PPI network, CDK2 and CDK4 had higher degrees. TFs ATF2 and E2F4 and miRNA miR-16 were predicted for the DEGs. Moreover, qRT-PCR analysis confirmed that SRR and KCNJ4 were significantly upregulated.These genes might affect the roles of DEX in the birth weight of infant, and might be promising therapeutic targets for reducing the side effects of DEX.


Asunto(s)
Peso al Nacer/genética , Dexametasona/efectos adversos , Glucocorticoides/efectos adversos , Análisis de Secuencia de ARN , Trofoblastos/efectos de los fármacos , Factor de Transcripción Activador 2/efectos de los fármacos , Peso al Nacer/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Factor de Transcripción E2F4/efectos de los fármacos , Femenino , Humanos , Recién Nacido , MicroARNs/efectos de los fármacos , N-Acetilgalactosaminiltransferasas/efectos de los fármacos , Placenta/citología , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Polipéptido N-Acetilgalactosaminiltransferasa
5.
Br J Cancer ; 117(8): 1154-1163, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-28873083

RESUMEN

BACKGROUND: Overexpression of epidermal growth factor receptor (EGFR) occurs in approximately 90% of head and neck squamous cell carcinoma (HNSCC), and is correlated with poor prognosis. Thus, targeting EGFR is a promising strategy for treatment of HNSCC. Several small molecule EGFR inhibitors have been tested in clinical trials for treatment of HNSCC, but none of them are more effective than the current chemotherapeutic drugs. Thus, it is urgently needed to develop novel EGFR inhibitors for HNSCC treatment. METHODS: By screening an in-house focused library containing approximately 650 000 known kinase inhibitors and kinase inhibitor-like compounds containing common kinase inhibitor core scaffolds, we identified SKLB188 as a lead compound for inhibition of EGFR. The anticancer effects of SKLB188 on HNSCC cells were investigated by in vitro cell growth, cell cycle and apoptosis assays, as well as in vivo FaDu xenograft mouse model. Molecular docking, in vitro kinase profiling and western blotting were performed to characterise EGFR as the molecular target. RESULTS: SKLB188 inhibited HNSCC cell proliferation by inducing G1 cell cycle arrest, which was associated with downregulating the expression of Cdc25A, cyclins D1/A and cyclin-dependent kinases (CDK2/4), and upregulating the expression of cyclin-dependent kinase (CDK) inhibitors (p21Cip1 and p27Kip1), leading to decreased phosphorylation of Rb. SKLB188 also induced caspase-dependent apoptosis of HNSCC cells by downregulating the expression of Mcl-1 and survivin. Molecular docking revealed that SKLB188 could bind to the kinase domain of EGFR through hydrogen bonds and hydrophobic interactions. In vitro kinase assay showed that SKLB188 inhibited the activity of a recombinant human EGFR very potently (IC50=5 nM). Western blot analysis demonstrated that SKLB188 inhibited the phosphorylation of EGFR and its downstream targets, extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) and Akt in the cells. In addition, SKLB188 dose-dependently inhibited FaDu xenograft growth in nude mice, and concurrently inhibited the phosphorylation of Erk1/2 and Akt in the tumours. CONCLUSIONS: SKLB188 potently inhibits the growth of HNSCC cells in vitro and in vivo by targeting EGFR signalling. The results provide a basis for further clinical investigation of SKLB188 as a targeted therapy for HNSCC. Our findings may open a new avenue for development of novel EGFR inhibitors for treatment of HNSCC and other cancers.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/metabolismo , Purinas/farmacología , Animales , Western Blotting , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Receptores ErbB/metabolismo , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Ratones , Ratones Desnudos , Simulación del Acoplamiento Molecular , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/efectos de los fármacos , Fosfatasas cdc25/metabolismo
6.
Asian Pac J Cancer Prev ; 15(8): 3651-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24870773

RESUMEN

Hepatocellular carcinoma (HCC) has a relatively higher incidence in many countries of Asia. Globally, HCC has a high fatality rate and short survival. Epirubicin, a doxorubicin analogue, may be administered alone or in combination with other agents to treat primary liver cancer and metastatic diseases. However, the toxic effects of epirubicin to normal tissues and cells have been one of the major obstacles to successful cancer chemotherapy. Here, we investigated the effects of epirubicin in combination with kappa-selenocarrageenan on mice with H22 implanted tumors and HepG-2 cell proliferation, immune organ index, morphology, cell cycle and related protein expressions in vivo and in vitro with sequential drug exposure. The inhibitory rate of tumor growth in vivo was calculated. Drug sensitivity was measured by MTT assay, and the King's principle was used to evaluate the interaction of drug combination. Morphological changes were observed by fluorescent microscopy. Cell cycle changes were analyzed by flow cytometry. Expression of cyclin A, Cdc25A and Cdk2 were detected by Western blotting. In vivo results demonstrated that the inhibitory rate of EPI combined with KSC was higher than that of KSC or EPI alone, and the Q value indicated an additive effect. In addition, KSC could significantly raise the thymus and spleen indices of mice with H22 implanted tumors. In the drug sensitivity assay in vitro, exposure to KSC and EPI simultaneously was more effective than exposure sequentially in HepG-2 cells, while exposure to KSC prior to EPI was more effective than exposure to EPI prior to KSC. Q values showed an additive effect in the simultaneous group and antagonistic effects in the sequential groups. Morphological analysis showed similar results to the drug sensitivity assay. Cell cycle analysis revealed that exposure to KSC or EPI alone arrested the cells in S phase in HepG-2 cells, exposure to KSC and EPI simultaneously caused accumulation in the S phase, an effect caused by either KSC or EPI. Expression of cyclin A, Cdc25A and Cdk2 protein was down-regulated following exposure to KSC and EPI alone or in combination, exposure to KSC and EPI simultaneously resulting in the lowest values. Taken together, our findings suggest that KSC in combination with EPI might have potential as a new therapeutic regimen against HCC.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Carcinoma Hepatocelular , Carragenina/farmacología , Epirrubicina/farmacología , Neoplasias Hepáticas , Compuestos de Organoselenio/farmacología , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Esquema de Medicación , Células Hep G2 , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/efectos de los fármacos , Fosfatasas cdc25/metabolismo
7.
Toxicol Appl Pharmacol ; 272(1): 12-20, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707764

RESUMEN

Licorice extract which is used as a natural sweetener has been shown to possess inhibitory effects against prostate cancer, but the mechanisms responsible are poorly understood. Here, we report a compound, isoangustone A (IAA) in licorice that potently suppresses the growth of aggressive prostate cancer and sought to clarify its mechanism of action. We analyzed its inhibitory effects on the growth of PTEN-deleted human prostate cancer cells, in vitro and in vivo. Administration of IAA significantly attenuated the growth of prostate cancer cell cultures and xenograft tumors. These effects were found to be attributable to inhibition of the G1/S phase cell cycle transition and the accumulation of p27(kip1). The elevated p27(kip1) expression levels were concurrent with the decrease of its phosphorylation at threonine 187 through suppression of CDK2 kinase activity and the reduced phosphorylation of Akt at Serine 473 by diminishing the kinase activity of the mammalian target of rapamycin (mTOR). Further analysis using recombinant proteins and immunoprecipitated cell lysates determined that IAA exerts suppressive effects against CDK2 and mTOR kinase activity by direct binding with both proteins. These findings suggested that the licorice compound IAA is a potent molecular inhibitor of CDK2 and mTOR, with strong implications for the treatment of prostate cancer. Thus, licorice-derived extracts with high IAA content warrant further clinical investigation for nutritional sources for prostate cancer patients.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Isoflavonas/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Serina-Treonina Quinasas TOR/efectos de los fármacos , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Activación Enzimática/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Ácido Glicirrínico/farmacología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Indicadores y Reactivos , Masculino , Ratones , Ratones Endogámicos BALB C , Fosforilación , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Int J Urol ; 19(7): 662-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22462738

RESUMEN

OBJECTIVE: Capsaicin is a specialized agonist of transient receptor potential vanilloid type 1 Ca2(+) channel, a member of the vanilloid receptor family of cation channels. We aimed to investigate the effects of capsaicin on the proliferation and cell death of human bladder cancer cells. METHODS: Human bladder cancer cell line 5637 was cultured and the expression of transient receptor potential vanilloid type 1 verified by immunofluorescence and Western blot. Cells were given different disposals (different capsaicin concentration with/without pre-treating with capsazepine; capsazepine, acting as a competitive antagonist of capsaicin) to observe cell viability, cell cycle and cell death by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay and flow cytometry. The apoptosis indexes, such as intracellular production of reactive oxygen species and mitochondrial membrane potential were assessed to elucidate the potential mechanism of capsaicin effects in the cells. RESULTS: Capsaicin decreased the viability of 5637 cells in a dose-dependent way. The flow cytometry outcome showed that capsaicin blocked the cell cycle in the G0/G1 period. The Western blot of cyclin-dependent-kinase involved in G1/S transfer verified this. Meanwhile, increased reactive oxygen species production and decreased mitochondrial membrane potential were detected in capsaicin-treated groups. CONCLUSIONS: Capsaicin induces cell death through increased reactive oxygen species and decreased mitochondrial membrane potential. Furthermore, capsaicin inhibits the proliferation of 5637 bladder carcinoma cells by cycle arrest with the inhibition of CDK2, CDK4 and CDK6.


Asunto(s)
Capsaicina/farmacología , Carcinoma/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPV/metabolismo
9.
Anticancer Drugs ; 23(4): 370-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22185819

RESUMEN

Artemisinin, a sesquiterpene phytolactone derived from Artemisia annua, is a potent antimalarial compound with promising anticancer properties, although the mechanism of its anticancer signaling is not well understood. Artemisinin inhibited proliferation and induced a strong G1 cell cycle arrest of cultured MCF7 cells, an estrogen-responsive human breast cancer cell line that represents an early-stage cancer phenotype, and effectively inhibited the in-vivo growth of MCF7 cell-derived tumors from xenografts in athymic nude mice. Artemisinin also induced a growth arrest of tumorigenic human breast cancer cell lines with preneoplastic and late stage cancer phenotypes, but failed to arrest the growth of a nontumorigenic human mammary cell line. Concurrent with the cell cycle arrest of MCF7 cells, artemisinin selectively downregulated the transcript and protein levels of the CDK2 and CDK4 cyclin-dependent kinases, cyclin E, cyclin D1, and the E2F1 transcription factor. Analysis of CDK2 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK2 gene expression was accounted for by the loss of CDK2 promoter activity. Chromatin immunoprecipitation revealed that artemisinin inhibited E2F1 interactions with the endogenous MCF7 cell CDK2 and cyclin E promoters. Moreover, constitutive expression of exogenous E2F1 prevented the artemisinin-induced cell cycle arrest and downregulation of CDK2 and cyclin E gene expression. Taken together, our results demonstrate that the artemisinin disruption of E2F1 transcription factor expression mediates the cell cycle arrest of human breast cancer cells and represents a critical transcriptional pathway by which artemisinin controls human reproductive cancer cell growth.


Asunto(s)
Antineoplásicos/farmacología , Artemisininas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Factor de Transcripción E2F1/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Factor de Transcripción E2F1/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Chem Inf Model ; 52(2): 373-9, 2012 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-22148673

RESUMEN

A case study is presented illustrating the design of a focused CDK2 library. The scaffold of the library was detected by a feature trees search in a fragment space based on reactions from combinatorial chemistry. For the design the software LoFT (Library optimizer using Feature Trees) was used. The special feature called FTMatch was applied to restrict the parts of the queries where the reagents are permitted to match. This way a 3D scoring function could be simulated. Results were compared with alternative designs by GOLD docking and ROCS 3D alignments.


Asunto(s)
Técnicas Químicas Combinatorias , Diseño de Fármacos , Bibliotecas de Moléculas Pequeñas , Programas Informáticos , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Humanos
11.
Toxicology ; 276(2): 103-9, 2010 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-20637255

RESUMEN

Mechanisms of hepatocyte proliferation triggered by tissue loss are distinguishable from those that promote proliferation in the intact liver in response to mitogens. Previous studies demonstrate that exogenous activation of the aryl hydrocarbon receptor (AhR), a soluble ligand-activated transcription factor in the basic helix-loop-helix family of proteins, suppresses compensatory liver regeneration elicited by surgical partial hepatectomy. The goal of the present study was to determine how AhR activation modulates hepatocyte cell cycle progression in the intact liver following treatment with the hepatomitogen, 1,4-bis[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP). Mice were pretreated with the exogenous AhR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) 24h prior to treatment with TCPOBOP (3 mg/kg).). In contrast to the suppressive effects of AhR activation observed during compensatory regeneration, TCDD pretreatment resulted in a 30-50% increase in hepatocyte proliferation in the intact liver of TCPOBOP-treated mice. Although pretreatment with TCDD suppressed CDK2 kinase activity and increased the association of CDK2 with negative regulatory proteins p21Cip1 and p27Kip1, a corresponding increase in CDK4/cyclin D1 association and CDK4 activity which culminated in enhanced phosphorylation of retinoblastoma protein, consistent with the increased proliferative response. These findings are in stark contrast to previous observations that the activated AhR can suppress hepatocyte proliferation in vivo and reveal a new complexity to AhR-mediated cell cycle control.


Asunto(s)
Hiperplasia/inducido químicamente , Hígado/efectos de los fármacos , Dibenzodioxinas Policloradas/farmacología , Piridinas/toxicidad , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/efectos de los fármacos , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo
12.
Environ Toxicol ; 24(3): 243-58, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18683188

RESUMEN

Humic acid (HA) in well water used by the inhabitants for drinking is one of the possible etiological factors for Blackfoot disease (BFD). In this study, the ability of HA to inhibit cell cycle progression and induce apoptosis in cultured smooth muscle cells (SMCs; A7r5) was investigated. Treatment of the SMCs at various HA concentrations (25-200 microg/mL) resulted in sequences of events marked by apoptosis, as shown by loss of cell viability, morphology change, and internucleosomal DNA fragmentation. HA-induced apoptotic cell death that is associated with loss of mitochondrial membrane potential (Delta Psi m), cytochrome c translocation, caspase-3, -8, and -9 activation, poly ADP-ribose polymerase (PARP) degradation, dysregulation of Bcl-2 and Bax, and upregulation of p53 and phospholyrated p53 (p-p53) in SMCs. Flow cytometry analysis demonstrated that HA blocked cell cycle progress in the G1 phase in SMCs. This blockade of cell cycle was associated with reduced amounts of cyclin D1, CDK4, cyclin E, CDK2, and hyperphosphorylated retinoblastoma protein (pRb) in a time-dependent manner. Apparent DNA strand breaks (DNA damage) were also detected in a dose-dependent manner using Single-cell gel electrophoresis assay (comet assay). Furthermore, HA induced dose-dependent elevation of reactive oxygen species (ROS) level in SMCs, and antioxidant vitamin C and Trolox effectively suppressed HA-induced DNA damage and dysregulation of Bcl-2/Bax. Our findings suggest that HA-induced DNA damage, cell cycle arrest, and apoptosis in SMCs may be an underlying mechanisms for the atherosclerosis and thrombosis observed in the BFD endemic region.


Asunto(s)
Apoptosis , Fase G1/efectos de los fármacos , Sustancias Húmicas/toxicidad , Músculo Liso Vascular/efectos de los fármacos , Animales , Caspasas/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo XI/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/genética , Ciclina E/efectos de los fármacos , Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/genética , Citocromos c/metabolismo , Fragmentación del ADN , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Proteína de Retinoblastoma/metabolismo , Proteína X Asociada a bcl-2/metabolismo
13.
Br J Dermatol ; 160(2): 442-5, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19016696

RESUMEN

BACKGROUND: The enzyme mammalian target of rapamycin (mTOR) integrates many different cellular signals to control cell growth and proliferation, protein synthesis and breakdown, and other processes. Dysregulation of mTOR is implicated in a range of human diseases, including cancers and cardiovascular disorders. To date, there has been no report on the expression of protein kinase B (AKT)/mTOR cell signalling in epidermal tumours. OBJECTIVES: This study was designed to investigate the activation of the mTOR signalling pathway in epidermal tumours and to correlate this with cyclin-dependent kinase 2 (CDK2) expression. METHODS: Immunohistological staining was performed with phosphorylated (p-) AKT, p-mTOR, p-4E-binding protein 1 (p-4EBP1), p-ribosomal protein S6 (p-S6), p-p70 ribosomal protein S6 kinase 1 (p-p70S6K1) and CDK2 in 15 cases each of seborrhoeic keratosis, actinic keratosis, keratoacanthoma and Bowen's disease (BD), and 25 cases of squamous cell carcinoma (SCC). Fifteen normal skin (NS) samples served as control. RESULTS: Among 85 tumours, 40 (47%) were positive for p-AKT, 31 (36%) for p-mTOR, 44 (52%) for p-4EBP1, 38 (45%) for p-S6, and 39 (46%) for p-p70S6K1. CDK2 immunostaining was positive in all cases of SCC and BD, and in 67% of benign tumours. All of these markers were stained much more frequently in malignant tumours than in benign tumours or NS. p-AKT, p-mTOR, p-4EBP1, p-p70S6K1 and p-S6 each showed high correlation with CDK2. CONCLUSIONS: Constitutive activation of the AKT/mTOR pathway was frequent in epidermal tumours, especially in malignant tumours. Activation was highly correlated with CDK2 expression, suggesting that the AKT/mTOR pathway may induce the malignant transition through CDK2 in epidermal tumours.


Asunto(s)
Enfermedad de Bowen/metabolismo , Carcinoma de Células Escamosas/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Queratosis Actínica/patología , Sirolimus/farmacología , Neoplasias Cutáneas/metabolismo , Carcinoma de Células Escamosas/patología , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Epidermis/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
14.
Neurosci Lett ; 450(3): 347-50, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19103257

RESUMEN

The extent to which neurons proceed into the cell cycle and the mechanisms whereby cell cycle re-entry leads to apoptosis vary in response to agonists. We previously showed upregulation of early G1 regulators in thrombin-treated neurons yet neurons did not proceed to S phase but to apoptosis. The objective of this study is to explore mechanisms which might prevent S phase entry and promote apoptosis in thrombin-treated neurons. Cultured rat brain neurons are exposed to thrombin (200nM) for 30min to 4.5h and the expression of cyclin C, cyclin dependent kinases (cdk1, cdk2, cdk3, cdk8) and the cell cycle inhibitor p27 assessed. Our data show a simultaneous decrease of both cyclin C and cdk3 proteins soon after thrombin treatment. The decrease in cyclin C also correlates with decreases in cdk1 and cdk2, at both mRNA and protein levels. There is no change in expression of cdk8 or the cell cycle inhibitor p27 in response to thrombin treatment. These results suggest that decreases in G1-S regulators cyclin C and cdks 3, cdk2 and cdk1 in response to thrombin could make conditions unfavorable for S phase entry and favor neuronal apoptosis.


Asunto(s)
Apoptosis/fisiología , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Neuronas/enzimología , Trombina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteína Quinasa CDC2/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Células Cultivadas , Ciclina C , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 3 Dependiente de Ciclina , Quinasa 8 Dependiente de Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/efectos de los fármacos , Ciclinas/metabolismo , Fase G1/efectos de los fármacos , Fase G1/fisiología , Neuronas/efectos de los fármacos , Ratas , Fase S/efectos de los fármacos , Fase S/fisiología , Trombina/metabolismo
15.
Mol Nutr Food Res ; 52(11): 1281-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18686292

RESUMEN

Methylseleninic acid (MSeA) is a synthetic organoselenium form known to be effective against mammary carcinogenesis in vivo. Using the synchronized mouse mammary epithelial tumor cell (TM6) model, we have previously shown that 5 microM MSeA significantly inhibits cell growth and induces a reversible growth arrest in the G1 phase. In the present study, we examined the effects of MSeA on Rb, cyclin dependent kinase 2 (cdk2), cdk4, cyclin E and cyclin D1. Growth arrest of cells was accompanied by a reduction in total cdk2 kinase and cyclin E-associated cdk2 kinase activities. The p27 levels associated with cdk2 were elevated during the cell cycle. In addition, growth inhibition correlated with a relative increase in the hypophosphorylated form of Rb in MSeA-treated cells and Egr1 was elevated in MSeA-treated cells. The Kinetworks Protein Kinase Screen (KPKS 1.0) was used to examine 75 protein kinases. MSeA treatment resulted in differential expression of several protein-serine/threonine kinases, protein-tyrosine kinases and protein-threonine/tyrosine kinases. Some of these kinases are being reported for the first time as being altered by MSeA. The outcome of these experiments will be of significance since these kinases are known to be involved in survival and/or apoptotic pathways of tumor cells.


Asunto(s)
Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/prevención & control , Compuestos de Organoselenio/uso terapéutico , Animales , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Replicación del ADN/efectos de los fármacos , Fase G1/efectos de los fármacos , Neoplasias Mamarias Animales/enzimología , Neoplasias Mamarias Animales/genética , Ratones , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo
16.
Toxicology ; 250(1): 39-46, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18577415

RESUMEN

In the present study, we investigated the effects of manganese chloride (MnCl2) on cell cycle progression in A549 cells used as a model of Mn-induced lung toxicity. Cells were treated with various concentrations of MnCl2 (0, 0.01, 0.1, 0.5, 1.0 or 2.0 mM) for 24, 48 or 72 h. Cell proliferation was determined with MTT assay and mitotic index measurement and apoptosis was measured by flow cytometer. The results showed that MnCl2 inhibited A549 cells proliferation in a dose- and time-dependent manner, and induced apoptosis in A549 cells. When G0/G1 cells obtained by serum starvation were incubated with 0.5 mM of MnCl2 in the presence of 10% serum for several time intervals, the disruption of cell cycle progression was observed. The G0/G1 arrest was induced by MnCl2 treatment at 16 h and the arrest maintained for 8 h. Following the G0/G1 arrest, MnCl2 blocked the cells at S phase at 28 h and the S phase arrest maintained for at least 4 h. And moreover, proteasome inhibitor MG132 was able to prolong the duration of G0/G1 arrest induced by MnCl2 treatment. Results of western blotting assay revealed that cellular Cdk4, Cdk2 and phospho-Cdk2 (Thr160) levels decreased in manganese-treated cells at both 20 and 28 h. In addition, the decreasing of Cyclin A level and the increasing of p53 and WAF1/p21 were also induced by MnCl2 treatment at 20 h. The expression of Cyclin D1, Cyclin E and Cdc25A proteins was not altered in manganese-treated cells at both 20 and 28 h. Our results indicate that MnCl2 orderly induces G0/G1 and S phase arrest in A549 cells, the decreasing of Cdk4, Cdk2 and Cyclin A, and the increasing of p53 and Cdks inhibitor WAF1/p21 might be responsible for the G0/G1 arrest, and the decreasing of Cdk4 and Cdk2 levels for the S phase arrest.


Asunto(s)
Cloruros/toxicidad , Fase G1/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase S/efectos de los fármacos , Adenocarcinoma/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cloruros/administración & dosificación , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Compuestos de Manganeso/administración & dosificación , Índice Mitótico , Factores de Tiempo
17.
Bioorg Med Chem Lett ; 17(22): 6216-9, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17904366
19.
Virology ; 367(1): 1-9, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17568647

RESUMEN

Epidemiological studies have demonstrated that 15 different mucosal human papillomavirus (HPV) types of the genus alpha of the HPV phylogetic tree are classified as high risk for cervical cancer development. Three additional HPV types of the same genus, HPV26, 53 and 66, are classified as probable high-risk types. In this study, we have characterized the biological properties of the E7 oncoproteins from these three HPV types. All of the corresponding E7 proteins were able to associate with retinoblastoma protein (pRb) and up-regulated the expression of several positive cell cycle regulators, i.e. CDK2, cyclin A and cylin E. However, HPV26 E7 appears to be more efficient than HPV53 and 66 E7 in up-regulating the transcription of cyclin A. Unlike E7 from the high-risk type HPV16 protein, HPV26, 53 and 66 did not efficiently promote pRb degradation. In addition, E7 from these viruses was able to promote proliferation of primary human keratinocytes and circumvent G1 arrest imposed by overexpression of p16(INK4a), but with less efficiency than the high-risk HPV16 E7. Together, our data show that in vitro properties of these E7 proteins correlate with the epidemiological classification of HPV26, 53 and 66 as HPV types with an intermediate risk for cervical cancer development.


Asunto(s)
Alphapapillomavirus/clasificación , Alphapapillomavirus/patogenicidad , Transformación Celular Neoplásica , Queratinocitos/virología , Proteínas E7 de Papillomavirus/metabolismo , Alphapapillomavirus/metabolismo , Línea Celular , Línea Celular Transformada , Células Cultivadas , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fase G1/efectos de los fármacos , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/farmacología , Infecciones por Papillomavirus/virología , Proteína de Retinoblastoma/metabolismo , Factores de Riesgo , Regulación hacia Arriba , Neoplasias del Cuello Uterino/virología
20.
Oncogene ; 26(24): 3521-31, 2007 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-17173073

RESUMEN

Molecular therapeutics is a recognized promising approach for melanoma, but relevant target genes remain elusive. We report that overload of the recently cloned H11/HspB8 induces apoptosis in 55% of examined melanoma cultures. Apoptosis was determined by activation of caspases-9 and -3 and terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL), and was not seen in normal melanocytes. It was associated with H11/HspB8 complexation with transforming growth factor-beta-activated kinase (TAK) 1 and activation of TAK1 and p38 mitogen activated protein 3 kinases. TAK1 was not bound, nor activated by the H11/HspB8 mutant W51C, which has dominant antiapoptotic activity. beta-Catenin was phosphorylated by activated TAK1, inhibiting its nuclear accumulation and mictophthalmia-associated transcription factor and cyclin dependent kinase 2 expression. The dominant-negative TAK1 mutant K63W inhibited beta-catenin phosphorylation and caspase activation. The data indicate that H11/HspB8 overload causes melanoma growth arrest and apoptosis through TAK1 activation and suggest that H11/HspB8 is a promising molecular therapy target.


Asunto(s)
Apoptosis/fisiología , Proteínas de Choque Térmico/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Melanoma/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Apoptosis/efectos de los fármacos , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Núcleo Celular/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Doxorrubicina/farmacología , Activación Enzimática , Proteínas de Choque Térmico/genética , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/efectos de los fármacos , Factor de Transcripción Asociado a Microftalmía/metabolismo , Chaperonas Moleculares , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Células Tumorales Cultivadas , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...