Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Mini Rev Med Chem ; 21(7): 792-802, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33238842

RESUMEN

CDATA[The inherited mutations and underexpression of BRCA1 in sporadic breast cancers resulting in the loss or functional inactivation of BRCA1 may contribute to a high risk of breast cancer. Recent researchers have identified small molecules (BRCA1 mimetics) that fit into a BRCA1 binding pocket within Estrogen Receptor alpha (ERα), mimic the ability of BRCA1 to inhibit ERα activity, and overcome antiestrogen resistance. Studies indicate that most of the BRCA1 breast cancer cases are associated with p53 mutations. It indicates that there is a potential connection between BRCA1 and p53. Most p53 mutations are missense point mutations that occur in the DNA-binding domain. Structural studies have demonstrated that mutant p53 core domain misfolding, especially p53-R175H, is reversible. Mutant p53 reactivation with a new class of zinc metallochaperones (ZMC) restores WT p53 structure and functions by restoring Zn2+ to Zn2+ deficient mutant p53. Considering the role of WT BRCA1 and reactivation of p53 in tumor cells, our hypothesis is to target both tumor suppressor proteins by a novel biomolecule (ZMC). Since both proteins are present in the same cell and are functionally inactive, this state may be a novel efficacious therapeutic regime for breast cancer therapy. In addition, we propose to use Albumin Nanovector (ANV) formulation for target drug release.


Asunto(s)
Albúminas/química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Chaperonas Moleculares/farmacología , Nanopartículas/química , Zinc/farmacología , Antineoplásicos/química , Ciclina D/antagonistas & inhibidores , Ciclina D/metabolismo , Portadores de Fármacos/química , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Chaperonas Moleculares/química , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Zinc/química
3.
Clin Transl Oncol ; 23(3): 612-619, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32710211

RESUMEN

BACKGROUND: Glioblastoma (GB) remains an incurable and deadly brain malignancy that often proves resistant to upfront treatment with temozolomide. Nevertheless, temozolomide remains the most commonly prescribed FDA-approved chemotherapy for GB. The DNA repair protein methylguanine-DNA methyl transferase (MGMT) confers resistance to temozolomide. Unsurprisingly temozolomide-resistant tumors tend to possess elevated MGMT protein levels or lack inhibitory MGMT promotor methylation. In this study, cultured human temozolomide resistance GB (43RG) cells were introduced to the MGMT inhibitor O6-benzylguanine combined with temozolomide and either LY2835219 (CDK 4/6 inhibitor) or LY2157299 (TGF-ßRI inhibitor) seeking to overcome GB treatment resistance. METHODS: Treatment effects were assessed using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, western blot, cell viability, and cell cycle progression. RESULTS: Our in vitro study demonstrated that sequential treatment of O6-Benzylguanine with either LY2385219 or LY2157299-enhanced temozolomide enhanced sensitivity in MGMT+ 43RG cells. Importantly, normal human neurons and astrocytes remained impervious to the drug therapies under these conditions. Furthermore, LY2835219 has additional anti-proliferative effects on cell cycling, including induction of an RB-associated G (1) arrest via suppression of cyclin D-CDK4/6-Rb pathway. LY2157299 enhances anti-tumor effect by disrupting TGF-ß-dependent HIF-1α signaling and by activating both Smad and PI3K-AKT pathways towards transcription of S/G2 checkpoints. CONCLUSION: This study establishes the groundwork for the development of a combinatorial pharmacologic approach by using either LY2385219 or LY2157299 inhibitor plus O6-Benzylguanine to augment temozolomide response in temozolomide-resistant GB cells.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Metilasas de Modificación del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Temozolomida/farmacología , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Aminopiridinas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Astrocitos/efectos de los fármacos , Bencimidazoles/farmacología , Neoplasias Encefálicas/enzimología , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclina D/antagonistas & inhibidores , Resistencia a Antineoplásicos/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular , Glioblastoma/enzimología , Guanina/análogos & derivados , Guanina/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Pirazoles/farmacología , Quinolinas/farmacología , Proteínas Smad/efectos de los fármacos
4.
Adv Cancer Res ; 148: 147-169, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32723562

RESUMEN

The mammalian cell cycle is driven by a complex of cyclins and their associated cyclin-dependent kinases (CDKs). Abnormal dysregulation of cyclin-CDK is a hallmark of cancer. D-type cyclins and their associated CDKs (CDK4 and CDK6) are key components of cell cycle machinery in driving G1 to S phase transition via phosphorylating and inactivating the retinoblastoma protein (RB). A body of evidence shows that the cyclin Ds-CDKs axis plays a critical role in cancer through various aspects, such as control of proliferation, senescence, migration, apoptosis, and angiogenesis. CDK4/6 dual-inhibitors show significant efficacy in pre-clinical or clinical cancer therapies either as single agents or in combination with hormone, chemotherapy, irradiation or immune treatments. Of note, as the associated partner of D-type cyclins, CDK6 shows multiple distinct functions from CDK4 in cancer. Depletion of the individual CDK may provide a therapeutic strategy for patients with cancer.


Asunto(s)
Ciclina D/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Humanos , Neoplasias/enzimología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología
5.
Cell Cycle ; 18(21): 3030-3043, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31544588

RESUMEN

This study was designed to purify molecules possess anti-cancer cell activity from the fruit body of Ganoderma leucocontextum. Bio-activity-guided purification and chromatographic separation of Ganoderma leucocontextum extract led to the enrichment of bioactive fractions and isolation of a single compound. The purified compound was identified as Ganoderiol F, which induced cancer cell death. In the in vivo experiments, we founded ethanol extract and ethyl acetate fraction inhibited tumor growth in the mice injected with 4T1 cells. We found that Ganoderiol F-mediated suppression of breast cancer cell viability occurred through cell cycle arrest. Ganoderiol F down-regulated expression of cyclin D, CDK4, CDK6, cyclin E and CDK2 and inhibited cell cycle progression arresting the cells in G1 phase. In addition, Ganoderiol F up-regulated pro-apoptotic Foxo3, down-regulated anti-apoptotic c-Myc, Bcl-2 and Bcl-w leading to apoptosis in human breast cancer cells MDA-MB-231. These results showed that c-Myc, cyclin D-CDK4/CDK6 and cyclin E-CDK2 are the central components of Ganoderiol F regulation of cell cycle progression. Hence Ganoderiol F may serve as a potential CDK4/CDK6 inhibitor for breast cancer therapy. Abbreviations: GLE: Ganoderma leucocontextum ethanol extract; GLEA: Ganoderma leucocontextum ethyl acetate fraction; GLPE: Ganoderma leucocontextum petroleum ether fraction; RP-HPLC: reversed-phase high-performance liquid chromatograph; DMEM: Dulbecco's modified Eagle's medium; FBS: fetal bovine serum; PAGE: polyacrylamide gel electrophoresis.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Triterpenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Extractos Celulares/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D/antagonistas & inhibidores , Ciclina E/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Femenino , Cuerpos Fructíferos de los Hongos/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Ganoderma/química , Humanos , Medicina Tradicional China , Ratones , Ratones Endogámicos BALB C
6.
Environ Toxicol ; 33(1): 41-51, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28941016

RESUMEN

Vitis thunbergii var. taiwaniana (VTT) is a wild grape native to Taiwan, belonging to the Vitaceae family and Vitis genus, and widely used as folk herbal medicine. It is traditionally used for the treatment of diarrhea, hypertension, neuroprotection, jaundice, and arthritis. We used the wild-collected VTT and sterilized them to establish the plant tissue culture, and then took the leaves for DNA sequencing to determine its original base. We use methanol to extract VTT in four different solvents: 1-butanol, n-hexane, ethyl acetate, and water. These four preliminary extracts were used to treat human prostate cancer DU145 cells in vitro. We use the flow cytometry to check the cell survival situation. Finally, we found the ethyl acetate layer roughing product (referred VTEA) in human prostate cancer apoptotic effects of cell line DU-145. In the present studies, we use the crude extract of VTT to examine whether or not it can induce apoptosis of DU145 cells in vitro. Viability assays for extracts of VTT treatment showed that it had dose-dependent effect on human prostate cancer DU145 cells. We also found that the extract of VTT induces time-dependent mitochondrial and intrinsic-dependent apoptosis pathways. The in vitro cytotoxic effects were investigated by cell cycle analysis and the determination of apoptotic DNA fragmentation in DU145 cells. The cell cycle analysis showed that extracts of VTT induced a significant increase in the number of cells in G0 /G1 phase. The extract of VTT induced chromatin changes and apoptosis of DU145 cells also were confirmed by DAPI and PI staining that were measured by fluorescence microscopy and flow cytometry, respectively. Finally, the expression of relevant proteins was analyzed by Western blot analysis. These results promoted us to further evaluate apoptosis associated proteins and elucidate the possible signal pathway in DU-145 cells after treated with the extract of VTT.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclina D/metabolismo , Ciclina E/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Vitis/química , Acetatos/química , Caspasas/metabolismo , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Ciclina D/antagonistas & inhibidores , Ciclina E/antagonistas & inhibidores , Fragmentación del ADN/efectos de los fármacos , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metanol/química , Microscopía Confocal , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Extractos Vegetales/análisis , Extractos Vegetales/química , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo , Taiwán , Vitis/metabolismo
7.
Biosci Biotechnol Biochem ; 80(6): 1095-106, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26924430

RESUMEN

Angiogenesis, neovascularization from pre-existing vessels, is a key step in tumor growth and metastasis, and anti-angiogenic agents that can interfere with these essential steps of cancer development are a promising strategy for human cancer treatment. In this study, we characterized the anti-angiogenic effects of Coptis japonica Makino extract (CJME) and its mechanism of action. CJME significantly inhibited the proliferation, migration, and invasion of vascular endothelial growth factor (VEGF)-stimulated HUVECs. Furthermore, CJME suppressed VEGF-induced tube formation in vitro and VEGF-induced microvessel sprouting ex vivo. According to our study, CJME blocked VEGF-induced cell cycle transition in G1. CJME decreased expression of cell cycle-regulated proteins, including Cyclin D, Cyclin E, Cdk2, and Cdk4 in response to VEGF. Taken together, the results of our study indicate that CJME suppresses VEGF-induced angiogenic events such as proliferation, migration, and tube formation via cell cycle arrest in G1.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Coptis/química , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Neovascularización Patológica/prevención & control , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/aislamiento & purificación , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D/antagonistas & inhibidores , Ciclina D/genética , Ciclina D/metabolismo , Ciclina E/antagonistas & inhibidores , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Extractos Vegetales/química , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/farmacología
8.
J Proteome Res ; 14(11): 4594-602, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26491887

RESUMEN

Many studies have shown the Na(+)/K(+)-ATPase (NKA) might be a potential target for anticancer therapy. Cardiac glycosides (CGs), as a family of naturally compounds, inhibited the NKA activity. The present study investigates the antitumor effect of ouabain and elucidates the pharmacological mechanisms of CG activity in liver cancer HepG2 cell using SILAC coupled to LC-MS/MS method. Bioinformatics analysis of 330 proteins that were changed in cells under treatment with 0.5 µmol/L ouabain showed that the biological processes are associated with an acute inflammatory response, cell cycle, oxidation reduction, chromosome segregation, and DNA metabolism. We confirmed that ouabain induced chromosome segregation disorder and S-cell cycle block by decreasing the expression of AURKA, SMC2, Cyclin D, and p-CDK1 as well as increasing the expression of p53. We found that the overexpression or inhibition of AURKA significantly reduced or enhanced the ouabain-mediated the anticancer effects. Our findings suggest that AURKA is involved in the anticancer mechanisms of ouabain in HepG2 cells.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasa A/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Ouabaína/farmacología , Fase S/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Animales , Aurora Quinasa A/genética , Aurora Quinasa A/metabolismo , Proteína Quinasa CDC2 , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Cromatografía Liquida , Segregación Cromosómica/efectos de los fármacos , Ciclina D/antagonistas & inhibidores , Ciclina D/genética , Ciclina D/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Femenino , Redes Reguladoras de Genes/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fase S/genética , Transducción de Señal , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Espectrometría de Masas en Tándem , Proteína p53 Supresora de Tumor/agonistas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Clin Cancer Res ; 21(13): 2905-10, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25941111

RESUMEN

Cancer cells bypass normal controls over mitotic cell-cycle progression to achieve a deregulated state of proliferation. The retinoblastoma tumor suppressor protein (pRb) governs a key cell-cycle checkpoint that normally prevents G1-phase cells from entering S-phase in the absence of appropriate mitogenic signals. Cancer cells frequently overcome pRb-dependent growth suppression via constitutive phosphorylation and inactivation of pRb function by cyclin-dependent kinase (CDK) 4 or CDK6 partnered with D-type cyclins. Three selective CDK4/6 inhibitors, palbociclib (Ibrance; Pfizer), ribociclib (Novartis), and abemaciclib (Lilly), are in various stages of development in a variety of pRb-positive tumor types, including breast cancer, melanoma, liposarcoma, and non-small cell lung cancer. The emerging, positive clinical data obtained to date finally validate the two decades-old hypothesis that the cyclin D-CDK4/6 pathway is a rational target for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos/uso terapéutico , Ciclina D/antagonistas & inhibidores , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Humanos , Terapia Molecular Dirigida , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal
10.
Pigment Cell Melanoma Res ; 27(2): 275-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24289491

RESUMEN

Expression profiling of microRNAs in melanoma lesional skin biopsies compared with normal donor skin biopsies, as well as melanoma cell lines compared with normal melanocytes, revealed that hsa-miR-206 was down-regulated in melanoma (-75.4-fold, P = 1.7 × 10(-4)). MiR-206 has been implicated in a large number of cancers, including breast, lung, colorectal, ovarian, and prostate cancers; however, its role in tumor development remains largely unknown, its biologic function is poorly characterized, and its targets affecting cancer cells are largely unknown. MiR-206 reduced growth and migration/invasion of multiple melanoma cell lines. Bioinformatics identified cell cycle genes CDK2, CDK4, Cyclin C, and Cyclin D1 as strong candidate targets. Western blots and 3'UTR reporter gene assays revealed that miR-206 inhibited translation of CDK4, Cyclin D1, and Cyclin C. Additionally, hsa-miR-206 transfection induced G1 arrest in multiple melanoma cell lines. These observations support hsa-miR-206 as a tumor suppressor in melanoma and identify Cyclin C, Cyclin D1, and CDK4 as miR-206 targets.


Asunto(s)
Ciclina D/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Melanoma/genética , Melanoma/patología , MicroARNs/metabolismo , Neoplasias Cutáneas/patología , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Biopsia , Carcinogénesis/genética , Carcinogénesis/patología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Movimiento Celular/genética , Proliferación Celular , Biología Computacional , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/enzimología , MicroARNs/genética , Persona de Mediana Edad , Modelos Biológicos , Datos de Secuencia Molecular , Invasividad Neoplásica , Biosíntesis de Proteínas , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Donantes de Tejidos
11.
Cell Rep ; 5(4): 961-73, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24268780

RESUMEN

The subgranular zone (SGZ) of the adult hippocampus contains a pool of quiescent neural progenitor cells (QNPs) that are capable of entering the cell cycle and producing newborn neurons. The mechanisms that control the timing and extent of adult neurogenesis are not well understood. Here, we show that QNPs of the adult SGZ express molecular-clock components and proliferate in a rhythmic fashion. The clock proteins PERIOD2 and BMAL1 are critical for proper control of neurogenesis. The absence of PERIOD2 abolishes the gating of cell-cycle entrance of QNPs, whereas genetic ablation of bmal1 results in constitutively high levels of proliferation and delayed cell-cycle exit. We use mathematical model simulations to show that these observations may arise from clock-driven expression of a cell-cycle inhibitor that targets the cyclin D/Cdk4-6 complex. Our findings may have broad implications for the circadian clock in timing cell-cycle events of other stem cell populations throughout the body.


Asunto(s)
Factores de Transcripción ARNTL/genética , Relojes Circadianos/genética , Hipocampo/crecimiento & desarrollo , Neurogénesis/genética , Proteínas Circadianas Period/genética , Factores de Transcripción ARNTL/biosíntesis , Animales , Ciclo Celular/genética , Proliferación Celular , Ciclina D/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales , Neuronas/citología , Proteínas Circadianas Period/biosíntesis , Regiones Promotoras Genéticas
12.
Cancer Cell ; 22(4): 438-51, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23079655

RESUMEN

D-cyclins represent components of cell cycle machinery. To test the efficacy of targeting D-cyclins in cancer treatment, we engineered mouse strains that allow acute and global ablation of individual D-cyclins in a living animal. Ubiquitous shutdown of cyclin D1 or inhibition of cyclin D-associated kinase activity in mice bearing ErbB2-driven mammary carcinomas triggered tumor cell senescence, without compromising the animals' health. Ablation of cyclin D3 in mice bearing Notch1-driven T cell acute lymphoblastic leukemias (T-ALL) triggered tumor cell apoptosis. Such selective killing of leukemic cells can also be achieved by inhibiting cyclin D associated kinase activity in mouse and human T-ALL models. Inhibition of cyclin D-kinase activity represents a highly-selective anticancer strategy that specifically targets cancer cells without significantly affecting normal tissues.


Asunto(s)
Ciclina D/fisiología , Neoplasias/etiología , Animales , Apoptosis , Puntos de Control del Ciclo Celular , Ciclina D/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Femenino , Humanos , Neoplasias Mamarias Experimentales/etiología , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Receptor ErbB-2/análisis
13.
Mol Vis ; 18: 758-64, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22509106

RESUMEN

PURPOSE: Nerve growth factor (NGF) plays an important role in promoting the healing of corneal wounds. However, the molecular mechanism by which NGF functions is unknown. We investigated the possible effects of NGF on phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) and mitogen activated protein kinase (MAPK)/extracellular signal-regulated kinase (Erk) pathways and cell growth in human corneal epithelial cells (HCECs). METHODS: We examined the effect of NGF on cell cycle and proliferation in HCECs by flow cytometry and cell proliferation assay, respectively. The levels of D-type cyclins in NGF-treated HCECs were determined by western blot. The tyrosine kinase A (TrkA), PI3K/Akt and MAPK/Erk pathways were then checked in cells stimulated with NGF for different time periods or cells undergoing a dose-dependent treatment. Furthermore, HCECs were treated with pathway inhibitors, LY294002 or PD98059, to confirm NGF-induced activations. RESULTS: We found that NGF had a positive effect on the growth of HCECs, and D-type cyclins, and it was correlated with the percentage of the G(1) to S progression. We also observed a time-dependent and dose-dependent effect of NGF on the PI3K/Akt and MAPK/Erk pathways. Furthermore, NGF affected cell cycle progression of HCECs by regulating cyclin D through Akt and Erk activation upon treatment with the pathway inhibitors, LY294002 for Akt or PD98059 for Erk pathways. CONCLUSIONS: NGF stimulation could promote cell proliferation and cell cycle progression of HCECs by activation of cyclin D via the PI3K/Akt and MAPK/Erk signaling pathways.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Córnea/citología , Células Epiteliales/citología , Factor de Crecimiento Nervioso/farmacología , Transducción de Señal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cromonas/farmacología , Córnea/efectos de los fármacos , Córnea/metabolismo , Ciclina D/antagonistas & inhibidores , Ciclina D/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Tiempo
14.
Cell Cycle ; 11(3): 554-68, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22293404

RESUMEN

Drosophila cyclinD (CycD) is the single fly ortholog of the mammalian cyclin D1 and promotes both cell cycle progression and cellular growth. However, little is known about how CycD promotes cell growth. We show here that CycD/Cdk4 hyperactivity leads to increased mitochondrial biogenesis (mitobiogenesis), mitochondrial mass, NRF-1 activity (Tfam transcript levels) and metabolic activity in Drosophila, whereas loss of CycD/Cdk4 activity has the opposite effects. Surprisingly, both CycD/Cdk4 addition and loss of function increase mitochondrial superoxide production and decrease lifespan, indicating that an imbalance in mitobiogenesis may lead to oxidative stress and aging. In addition, we provide multiple lines of evidence indicating that CycD/Cdk4 activity affects the hypoxic status of cells and sensitizes animals to hypoxia. Both mitochondrial and hypoxia-related effects can be detected at the global transcriptional level. We propose that mitobiogenesis and the hypoxic stress response have an antagonistic relationship, and that CycD/Cdk4 levels regulate mitobiogenesis contemporaneous to the cell cycle, such that only when cells are sufficiently oxygenated can they proliferate.


Asunto(s)
Envejecimiento , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Proteínas de Drosophila/metabolismo , Hipoxia , Mitocondrias/metabolismo , Complejos de ATP Sintetasa/metabolismo , Animales , Ciclina D/antagonistas & inhibidores , Ciclina D/genética , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/genética , ADN Mitocondrial/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Femenino , L-Lactato Deshidrogenasa/metabolismo , Masculino , Mitocondrias/genética , Factor 1 Relacionado con NF-E2/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Superóxidos/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
J Chem Inf Model ; 52(1): 76-83, 2012 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-22172011

RESUMEN

The cyclin dependent kinases (CDKs), each with their respective regulatory partner cyclin that are involved in the regulation of the cell cycle, apoptosis, and transcription, are potentially interesting targets for cancer therapy. The CDK6 complex with cyclin D (CDK6/cycD) drives cellular proliferation by phosphorylation of specific key target proteins. To understand the flavonoids that inhibit the CDK6/cycD functions, molecular dynamics simulations (MDSs) were performed on three inhibitors, fisetin (FST), apigenin (AGN), and chrysin (CHS), complexed with CDK6/cycD, including the two different binding orientations of CHS: FST-like (CHS_A) and deschloro-flavopiridol-like (CHS_B). For all three inhibitors, including both CHS orientations, the conserved interaction between the 4-keto group of the flavonoid and the backbone V101 nitrogen of CDK6 was strongly detected. The 3'- and 4'-OH groups on the flavonoid phenyl ring and the 3-OH group on the benzopyranone ring of inhibitor were found to significantly increase the binding and inhibitory efficiency. Besides the electrostatic interactions, especially through hydrogen bond formation, the van der Waals (vdW) interactions with the I19, V27, F98, H100, and L152 residues of CDK6 are also important factors in the binding efficiency of flavonoids against the CDK6/cycD complex. On the basis of the docking calculation and MM-PBSA method, the order of the predicted inhibitory affinities of these three inhibitors toward the CDK6/cycD was FST > AGN > CHS, which is in good agreement with the experimental data. In addition, CHS preferentially binds to the active CDK6 in a different orientation to FST and AGN but similar to its related analog, deschloro-flavopiridol. The obtained results are useful as the basic information for the further design of potent anticancer drugs specifically targeting the CDK6 enzyme.


Asunto(s)
Apigenina/química , Ciclina D/química , Quinasa 6 Dependiente de la Ciclina/química , Flavonoides/química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Apigenina/farmacología , Sitios de Unión , Cristalografía por Rayos X , Ciclina D/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Flavonoides/farmacología , Flavonoles , Humanos , Enlace de Hidrógeno , Conformación Molecular , Simulación de Dinámica Molecular , Piperidinas/química , Piperidinas/farmacología , Unión Proteica , Electricidad Estática , Termodinámica
16.
Eur J Med Chem ; 46(12): 5825-32, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22000924

RESUMEN

The design and synthesis of a small library of 4-aminopyrido[2,3-d]pyrimidine derivatives is reported. The potential activity of these compounds as CDK2/Cyclin A, CDK4/Cyclin D, EGFR and anti-tumor was evaluated by cytotoxicity studies in A431a, SNU638b, HCT116 and inhibition of CDK2-Cyclin A, CDK4/Cyclin D and EGFR enzyme activity in vitro. The anti-proliferative and CDK2-Cyclin A inhibitory activity of compounds 4c and 11a was significantly more active than roscovotine with IC(50) 0.3 and 0.09 µM respectively. Molecular modeling study, including fitting to a 3D-pharmacophore model, docking into cyclin dependant kinase2 (CDK2) active site and binding energy calculations were carried out and these studies suggested the same binding orientation inside the CDK2 binding pocket for these analogs compared to ATP.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Pirimidinas/química , Pirimidinas/farmacología , Antineoplásicos/síntesis química , Sitios de Unión , Línea Celular Tumoral , Ciclina A/antagonistas & inhibidores , Ciclina A/metabolismo , Ciclina D/antagonistas & inhibidores , Ciclina D/metabolismo , Quinasa 2 Dependiente de la Ciclina/química , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Pirimidinas/síntesis química
17.
Anticancer Res ; 31(9): 2821-32, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21868525

RESUMEN

Gallic acid (GA) induces apoptosis in different types of cancer cell lines. In this study, we investigate the apoptotic effects induced by GA in human promyelocytic leukemia HL-60 cells, and clarify the underlying mechanism. Our results showed that GA reduced the viability of HL-60 cells in a dose- and time-dependent manner. GA led to G(0)/G(1) phase arrest in HL-60 cells through promoting p21 and p27 and inhibiting the levels of cyclin D and cyclin E. GA caused DNA damage and fragmentation in HL-60 cells as assayed using DAPI staining and Comet assay. Flow cytometric analysis revealed that GA increased Ca(2+) levels and reduced the mitochondrial membrane potential (ΔΨ(m)) in HL-60 cells. Apoptotic protein expressions were determined by Western blotting. The results indicated that GA-mediated apoptosis of HL-60 cells mainly depended on mitochondrial pathway, by promoting the release of cytochrome c, apoptosis-inducing factor (AIF) and endonuclease G (Endo G) and by up-regulating the protein expression of Bcl-2-associated X protein (BAX), caspase-4, caspase-9 and caspase-3. In addition, GA also activated the death receptor-dependent pathway by enhancing the protein expressions of fatty acid synthase (FAS), FAS ligand (FASL), caspase-8 and BCL-2 interacting domain (BID). We determined the mRNA expression of the gene levels of these proteins by real-time PCR. The results showed that GA-mediated apoptosis of HL-60 cells mainly depended on up-regulation of the mRNA of caspase-8, caspase-9, caspase-3, AIF and Endo G. In conclusion, GA-induced apoptosis occurs through the death receptor and mitochondria-mediated pathways. The evaluation of GA as a potential therapeutic agent for treatment of leukemia seems warranted.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclina D/antagonistas & inhibidores , Ciclina E/antagonistas & inhibidores , Fase G1/efectos de los fármacos , Ácido Gálico/farmacología , Mitocondrias/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Secuencia de Bases , Ensayo Cometa , Cartilla de ADN , Citometría de Flujo , Células HL-60 , Humanos , Reacción en Cadena de la Polimerasa
18.
Cancer Biol Ther ; 11(7): 615-26, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21278493

RESUMEN

The c-Myc protein, encoded by c-myc gene, in its wild-type form can induce tumors with a high frequency and can induce massive programmed cell death (PCD) in most transgenic mouse models, with greater efficiency than other oncogenes. Evidence also indicates that c-Myc can cause proliferative inhibition, i.e. mitoinhibition. The c-Myc-induced PCD and mitoinhibition, which may be attributable to its inhibition of cyclin D1 and induction of p53, may impose a pressure of compensatory proliferation, i.e. regeneration, onto the initiated cells (cancer progenitor cells) that occur sporadically and are resistant to the mitoinhibition. The initiated cells can thus proliferate robustly and progress to a malignancy. This hypothetical thinking, i.e. the concurrent PCD and mitoinhibition induced by c-Myc can promote carcinogenesis, predicts that an optimal balance is achieved between cell death and ensuing regeneration during oncogenic transformation by c-Myc, which can better promote carcinogenesis. In this perspective, we summarize accumulating evidence and challenge the current model that oncoprotein induces carcinogenesis by promoting cellular proliferation and/or inhibiting PCD. Inspired by c-myc oncogene, we surmise that many tumor-suppressive or growth-inhibitory genes may also be able to promote carcinogenesis in a similar way, i.e. by inducing PCD and/or mitoinhibition of normal cells to create a need for compensatory proliferation that drives a robust replication of initiating cells.


Asunto(s)
Apoptosis/genética , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Reguladoras de la Apoptosis/metabolismo , Proliferación Celular , Ciclina D/antagonistas & inhibidores , Ciclina D/metabolismo , Humanos , Proteínas Oncogénicas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo
19.
Mol Pharmacol ; 79(4): 672-80, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21220410

RESUMEN

Amitriptyline is a classic tricyclic antidepressant (TCA) and has been used to treat the depression and anxiety of patients with cancer, but its relevance to cancer cell apoptosis is not known. In the present study, we demonstrated that amitriptyline inhibited cyclin D2 transactivation and displayed potential antimyeloma activity by inhibiting histone deacetylases (HDACs). Amitriptyline markedly decreased cyclin D2 promoter-driven luciferase activity, reduced cyclin D2 expression, and arrested cells at the G(0)/G(1) phase of the cell cycle. Amitriptyline-induced apoptosis was confirmed by Annexin V staining, and cleavage of caspase-3 and poly(ADP-ribose) polymerase-1. D-Cyclin expression is reported to be epigenetically regulated by histone acetylation. Thus, we examined the effects of amitriptyline on histone 3 (H3) acetylation and demonstrated that amitriptyline increased acetylation of H3 and expression of p27 and p21. Further studies indicated that amitriptyline interfered with HDAC function by down-regulation of HDAC3, -6, -7, and -8, but not HDAC2, and by interacting with HDAC7. Molecular docking analysis and molecular dynamics simulations revealed that amitriptyline bound to HDAC7 and formed strong van der Waals interactions with five residues of HDAC7, including Phe162, His192, Phe221, Leu293, and His326, thus inhibiting HDAC activity. Therefore, we found that amitriptyline inhibited cyclin D2 transactivation and HDAC activity and could be a promising treatment for multiple myeloma.


Asunto(s)
Amitriptilina/farmacología , Antidepresivos Tricíclicos/farmacología , Apoptosis/efectos de los fármacos , Ciclina D/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Mieloma Múltiple/metabolismo , Apoptosis/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Ciclina D/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología
20.
Blood ; 117(6): 1986-97, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21135258

RESUMEN

D-cyclins are universally dysregulated in multiple myeloma and frequently overexpressed in leukemia. To better understand the role and impact of dysregulated D-cyclins in hematologic malignancies, we conducted a high-throughput screen for inhibitors of cyclin D2 transactivation and identified 8-ethoxy-2-(4-fluorophenyl)-3-nitro-2H-chromene (S14161), which inhibited the expression of cyclins D1, D2, and D3 and arrested cells at the G(0)/G(1) phase. After D-cyclin suppression, S14161 induced apoptosis in myeloma and leukemia cell lines and primary patient samples preferentially over normal hematopoietic cells. In mouse models of leukemia, S14161 inhibited tumor growth without evidence of weight loss or gross organ toxicity. Mechanistically, S14161 inhibited the activity of phosphoinositide 3-kinase in intact cells and the activity of the phosphoinositide 3-kinases α, ß, δ, and γ in a cell-free enzymatic assay. In contrast, it did not inhibit the enzymatic activities of other related kinases, including the mammalian target of rapamycin, the DNA-dependent protein kinase catalytic subunit, and phosphoinositide-dependent kinase-1. Thus, we identified a novel chemical compound that inhibits D-cyclin transactivation via the phosphoinositide 3-kinase/protein kinase B signaling pathway. Given its potent antileukemia and antimyeloma activity and minimal toxicity, S14161 could be developed as a novel agent for blood cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Benzopiranos/farmacología , Ciclina D/antagonistas & inhibidores , Ciclina D/genética , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Activación Transcripcional/efectos de los fármacos , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Secuencia de Bases , Benzopiranos/química , Transporte Biológico Activo/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Cartilla de ADN/genética , Evaluación Preclínica de Medicamentos , Fase G1/efectos de los fármacos , Humanos , Células K562 , Leucemia/genética , Leucemia/patología , Ratones , Ratones SCID , Estructura Molecular , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Células 3T3 NIH , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...