Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Bioorg Med Chem ; 84: 117263, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37011445

RESUMEN

To search for potent CDK4/6 covalent inhibitors, total 14 compounds have been designed and synthesized by connecting different Michael-acceptor to the piperazine moiety of palbociclib. All the compounds displayed good antiproliferative activity against human hepatoma cell (HepG2), non-small cell lung cancer (A549), and breast cancer (MDA-MB-231 and MCF-7) cell lines. In particular, compound A4 showed the highest inhibitory activity to MDA-MB-231 and MCF-7 cells with IC50 values of 0.51 µM and 0.48 µM, respectively. More importantly, A4 also showed strong inhibition against MDA-MB-231/palbociclib cells, indicating that A4 could effectively avoid the resistance of palbociclib. In the enzyme test, A4 showed selective inhibitory activity against CDK4/6, with the IC50 value of 18 nM and 13 nM, respectively. It was also found that A4 could efficiently induce apoptosis and arrest the cell cycle at G0/G1 phase. Moreover, A4 could significantly decrease the phosphorylation level of CDK4 and CDK6. HPLC and molecular modeling studies suggested that A4 could form a covalent bond with the target protein.


Asunto(s)
Antineoplásicos , Resistencia a Antineoplásicos , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas , Línea Celular Tumoral , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/química , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Pulmonares/tratamiento farmacológico , Piperazinas/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/química , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Hepatocelular/tratamiento farmacológico
2.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34948218

RESUMEN

The G1 phase of cell cycle progression is regulated by Cyclin-Dependent Kinase 4 (CDK4) as well as Cyclin-Dependent Kinase 6 (CDK6), and the acivities of these enzymes are regulated by the catalytic subunit, cyclin D. Cell cycle control through selective pharmacological inhibition of CDK4/6 has proven to be beneficial in the treatment of estrogen receptor-positive (ER-positive) breast cancer, particularly improving the progression-free survival of patients. Thus, targeting specific inhibition on CDK4/6 is bound to increase therapeutic efficiency. This study aimed to obtain CDK4/6 inhibitors through a pharmacophore-based virtual screening of the ZINC15 purchasable compound database using the in silico method. The pharmacophore model was designed based on the FDA-approved cdk4/6 inhibitor structures, and molecular docking was performed to further screen the hit compounds obtained. A total of eight compounds were selected based on docking results and interactions with CDK4 and CDK6, using palbociclib as the reference drug. According to the results, the compounds of ZINC585292724 and ZINC585291674 were the best compounds based on free binding energy, as well as hydrogen bond stability, and, therefore, exhibit potential as starting points in the development of CDK4/6 inhibitors.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Piperazinas/química , Inhibidores de Proteínas Quinasas/química , Piridinas/química , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 6 Dependiente de la Ciclina/química , Humanos
3.
Molecules ; 26(8)2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33919867

RESUMEN

Novel 6-bromo-coumarin-ethylidene-hydrazonyl-thiazolyl and 6-bromo-coumarin-thiazolyl-based derivatives were synthesized. A quantitative structure activity relationship (QSAR) model with high predictive power r2 = 0.92, and RMSE = 0.44 predicted five compounds; 2b, 3b, 5a, 9a and 9i to have potential anticancer activities. Compound 2b achieved the best ΔG of -15.34 kcal/mol with an affinity of 40.05 pki. In a molecular dynamic study 2b showed an equilibrium at 0.8 Å after 3.5 ns, while flavopiridol did so at 0.5 Å after the same time (3.5 ns). 2b showed an IC50 of 0.0136 µM, 0.015 µM, and 0.054 µM against MCF-7, A-549, and CHO-K1 cell lines, respectively. The CDK4 enzyme assay revealed the significant CDK4 inhibitory activity of compound 2b with IC50 of 0.036 µM. The selectivity of the newly discovered lead compound 2b toward localization in tumor cells was confirmed by a radioiodination biological assay that was done via electrophilic substitution reaction utilizing the oxidative effect of chloramine-t. 131I-2b showed good in vitro stability up to 4 h. In solid tumor bearing mice, the values of tumor uptake reached a height of 5.97 ± 0.82%ID/g at 60 min p.i. 131I-2b can be considered as a selective radiotheranostic agent for solid tumors with promising anticancer activity.


Asunto(s)
Antineoplásicos/farmacología , Cumarinas/farmacología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Descubrimiento de Drogas , Radioisótopos de Yodo/química , Simulación de Dinámica Molecular , Relación Estructura-Actividad Cuantitativa , Células A549 , Secuencia de Aminoácidos , Animales , Antineoplásicos/química , Células CHO , Muerte Celular/efectos de los fármacos , Cumarinas/química , Cricetulus , Quinasa 2 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/metabolismo , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Células MCF-7 , Simulación del Acoplamiento Molecular , Distribución Tisular/efectos de los fármacos
4.
Int J Mol Sci ; 21(14)2020 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-32708403

RESUMEN

Chronic myeloid leukemia (CML) is a malignant tumor caused by the abnormal proliferation of hematopoietic stem cells. Among a new series of acridone derivatives previously synthesized, it was found that the methoxybenzyl 5-nitroacridone derivative 8q has nanomolar cytotoxicity in vitro against human chronic myelogenous leukemia K562 cells. In order to further explore the possible anti-leukemia mechanism of action of 8q on K562 cells, a metabolomics and molecular biology study was introduced. It was thus found that most of the metabolic pathways of the G1 phase of K562 cells were affected after 8q treatment. In addition, a concentration-dependent accumulation of cells in the G1 phase was observed by cell cycle analysis. Western blot analysis showed that 8q significantly down-regulated the phosphorylation level of retinoblastoma-associated protein (Rb) in a concentration-dependent manner, upon 48 h treatment. In addition, 8q induced K562 cells apoptosis, through both mitochondria-mediated and exogenous apoptotic pathways. Taken together, these results indicate that 8q effectively triggers G1 cell cycle arrest and induces cell apoptosis in K562 cells, by inhibiting the CDK4/6-mediated phosphorylation of Rb. Furthermore, the possible binding interactions between 8q and CDK4/6 protein were clarified by homology modeling and molecular docking. In order to verify the inhibitory activity of 8q against other chronic myeloid leukemia cells, KCL-22 cells and K562 adriamycin-resistant cells (K562/ADR) were selected for the MTT assay. It is worth noting that 8q showed significant anti-proliferative activity against these cell lines after 48 h/72 h treatment. Therefore, this study provides new mechanistic information and guidance for the development of new acridones for application in the treatment of CML.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteína de Retinoblastoma/metabolismo , Apoptosis/genética , Caspasas/metabolismo , Proliferación Celular/genética , Cromatografía Liquida , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/genética , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Espectrometría de Masas , Metabolómica , Simulación del Acoplamiento Molecular , Fosforilación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
5.
Molecules ; 25(9)2020 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-32349307

RESUMEN

On account of their overexpression in a wide range of human malignancies, cyclin-dependent kinases (CDKs) are among the most validated cancer targets, and their inhibition has been featured as a valuable strategy for anticancer drug discovery. In this study, a hybrid pharmacophore approach was adopted to develop two series of oxindole-indole conjugates (6a-i and 9a-f) and carbocycle-indole conjugates (11a,b) as efficient antitumor agents with potential inhibitory action toward CDK4. All oxindole-indole conjugates, except 6i, 9b, and 9c efficiently affected the growth of the human breast cancer MCF-7 (IC50: 0.39 ± 0.05-21.40 ± 1.58 µM) and/or MDA-MB-231 (IC50: 1.03 ± 0.04-22.54 ± 1.67 µM) cell lines, whereas bioisosteric replacement of the oxindole nucleus with indane or tetralin rings (compounds 11a,b) diminished the anti-proliferative activity. In addition, hybrids 6e and 6f displayed effective cell cycle disturbance and proapoptotic capabilities in MCF-7 cells. Furthermore, the efficient anti-proliferative agents towards MCF-7 and/or MDA-MB-231 cell lines (6a-h, 9a, and 9e) were investigated for their potential inhibitory action toward CDK4. Hybrids 6a and 6e displayed good CDK4 inhibitory activity with IC50s equal 1.82 and 1.26 µM, respectively. The molecular docking study revealed that oxindole moiety is implicated in two H-bonding interactions via both (NH) and (C=O) groups with the key amino acids Glu94 and Val96, respectively, whereas the indole framework is stably accommodated in a hydrophobic sub-pocket establishing hydrophobic interactions with the amino acid residues of Ile12, Val20, and Gln98 lining this sub-pocket. Collectively, these results highlighted hybrids 6a and 6e as good leads for further optimization as promising antitumor drugs toward breast malignancy and CDK inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Indoles/química , Oxindoles/química , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Sitios de Unión , Puntos de Control del Ciclo Celular/efectos de los fármacos , Simulación por Computador , Quinasa 4 Dependiente de la Ciclina/química , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Indoles/farmacología , Concentración 50 Inhibidora , Células MCF-7 , Simulación del Acoplamiento Molecular , Estructura Molecular , Oxindoles/farmacología , Inhibidores de Proteínas Quinasas/síntesis química , Relación Estructura-Actividad
6.
Nat Prod Res ; 34(19): 2715-2722, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30887847

RESUMEN

Fungal factories emerge as a promising source for the production of bioactive natural products. This study reports the isolation and structure elucidation of pulchranin A, for the first time, from an endophytic fungus (Aspergillus TRL1), which was cultured from Tabebuia rosea (Bignoniaceae) stems and identified by DNA ITS sequencing. Pulchranin A showed promising in-vitro cytotoxic effects against breast (MCF-7), liver (Hep-G2) and colorectal (HCT) cell lines, with IC50 values of 63, 80 and 91 µg/mL, respectively. In addition, it inhibited three cyclin-dependent kinases (CDK1, CDK2 and CDK4) in MCF-7 cells with IC50 values of 9.82, 15.6 and 2.7 µg/mL, respectively. Results were further supported by in-silico molecular docking of pulchranin A to CDK1, CDK2, and CDK4 crystal structures, where it demonstrated good interactions by H-bonding, hydrophobic and Pi-Pi interactions with different amino acid residues of these enzymes. Pulchranin A might be a potential CDK inhibitor in human breast cancer cells.[Figure: see text].


Asunto(s)
Antineoplásicos/farmacología , Aspergillus/química , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Guanidinas/química , Guanidinas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/química , Aspergillus/genética , Aspergillus/aislamiento & purificación , Bignoniaceae/microbiología , Proteína Quinasa CDC2/antagonistas & inhibidores , Proteína Quinasa CDC2/química , Proteína Quinasa CDC2/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , 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/química , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Endófitos/química , Células Hep G2 , Humanos , Enlace de Hidrógeno , Células MCF-7 , Simulación del Acoplamiento Molecular
7.
Science ; 366(6471)2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31831640

RESUMEN

The p27 protein is a canonical negative regulator of cell proliferation and acts primarily by inhibiting cyclin-dependent kinases (CDKs). Under some circumstances, p27 is associated with active CDK4, but no mechanism for activation has been described. We found that p27, when phosphorylated by tyrosine kinases, allosterically activated CDK4 in complex with cyclin D1 (CDK4-CycD1). Structural and biochemical data revealed that binding of phosphorylated p27 (phosp27) to CDK4 altered the kinase adenosine triphosphate site to promote phosphorylation of the retinoblastoma tumor suppressor protein (Rb) and other substrates. Surprisingly, purified and endogenous phosp27-CDK4-CycD1 complexes were insensitive to the CDK4-targeting drug palbociclib. Palbociclib instead primarily targeted monomeric CDK4 and CDK6 (CDK4/6) in breast tumor cells. Our data characterize phosp27-CDK4-CycD1 as an active Rb kinase that is refractory to clinically relevant CDK4/6 inhibitors.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Regulación Alostérica , Antineoplásicos/farmacología , Biocatálisis , Línea Celular Tumoral , Cristalografía por Rayos X , Ciclina D1/química , Quinasa 4 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Activación Enzimática , Humanos , Fosforilación , Conformación Proteica , Proteína de Retinoblastoma/metabolismo
8.
Mol Cell ; 74(4): 758-770.e4, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30982746

RESUMEN

The cyclin-dependent kinases Cdk4 and Cdk6 form complexes with D-type cyclins to drive cell proliferation. A well-known target of cyclin D-Cdk4,6 is the retinoblastoma protein Rb, which inhibits cell-cycle progression until its inactivation by phosphorylation. However, the role of Rb phosphorylation by cyclin D-Cdk4,6 in cell-cycle progression is unclear because Rb can be phosphorylated by other cyclin-Cdks, and cyclin D-Cdk4,6 has other targets involved in cell division. Here, we show that cyclin D-Cdk4,6 docks one side of an alpha-helix in the Rb C terminus, which is not recognized by cyclins E, A, and B. This helix-based docking mechanism is shared by the p107 and p130 Rb-family members across metazoans. Mutation of the Rb C-terminal helix prevents its phosphorylation, promotes G1 arrest, and enhances Rb's tumor suppressive function. Our work conclusively demonstrates that the cyclin D-Rb interaction drives cell division and expands the diversity of known cyclin-based protein docking mechanisms.


Asunto(s)
Proliferación Celular/genética , Ciclina D/genética , Mapas de Interacción de Proteínas/genética , Proteína de Retinoblastoma/genética , Ciclo Celular/genética , Proteína Sustrato Asociada a CrK/genética , Ciclina D/química , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/química , Quinasa 6 Dependiente de la Ciclina/genética , Ciclinas/genética , Fase G1/genética , Humanos , Simulación del Acoplamiento Molecular , Fosforilación/genética , Unión Proteica/genética , Conformación Proteica en Hélice alfa/genética , Proteína de Retinoblastoma/química , Proteína p107 Similar a la del Retinoblastoma/genética , Fase S/genética
9.
Mol Cancer Ther ; 18(4): 771-779, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30837298

RESUMEN

The interaction of a drug with its target is critical to achieve drug efficacy. In cases where cellular environment influences target engagement, differences between individuals and cell types present a challenge for a priori prediction of drug efficacy. As such, characterization of environments conducive to achieving the desired pharmacologic outcome is warranted. We recently reported that the clinical CDK4/6 inhibitor palbociclib displays cell type-specific target engagement: Palbociclib engaged CDK4 in cells biologically sensitive to the drug, but not in biologically insensitive cells. Here, we report a molecular explanation for this phenomenon. Palbociclib target engagement is determined by the interaction of CDK4 with CDKN2A, a physiologically relevant protein inhibitor of CDK4. Because both the drug and CDKN2A prevent CDK4 kinase activity, discrimination between these modes of inhibition is not possible by traditional kinase assays. Here, we describe a chemo-proteomics approach that demonstrates high CDK4 target engagement by palbociclib in cells without functional CDKN2A and attenuated target engagement when CDKN2A (or related CDKN2/INK4 family proteins) is abundant. Analysis of biological sensitivity in engineered isogenic cells with low or absent CDKN2A and of a panel of previously characterized cell lines indicates that high levels of CDKN2A predict insensitivity to palbociclib, whereas low levels do not correlate with sensitivity. Therefore, high CDKN2A may provide a useful biomarker to exclude patients from CDK4/6 inhibitor therapy. This work exemplifies modulation of kinase target engagement by endogenous proteinaceous regulators and highlights the importance of cellular context in predicting inhibitor efficacy.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Biomarcadores de Tumor/metabolismo , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/química , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/química , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Células MCF-7 , Mutación Missense , Proteínas del Tejido Nervioso/genética , Piperazinas/química , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Piridinas/química , Transfección
10.
Cell Chem Biol ; 26(2): 300-306.e9, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30595531

RESUMEN

The design of selective small molecules is often stymied by similar ligand binding pockets. Here, we report BSJ-03-123, a phthalimide-based degrader that exploits protein-interface determinants to achieve proteome-wide selectivity for the degradation of cyclin-dependent kinase 6 (CDK6). Pharmacologic CDK6 degradation targets a selective dependency of acute myeloid leukemia cells, and transcriptomics and phosphoproteomics profiling of acute degradation of CDK6 enabled dynamic mapping of its immediate role in coordinating signaling and transcription.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/metabolismo , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/química , Quinasa 6 Dependiente de la Ciclina/genética , Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ftalimidas/química , Ftalimidas/farmacología , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química
11.
J Mol Graph Model ; 82: 48-58, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29680736

RESUMEN

Cyclin Dependent Kinase 4 is a striking target for the proposal of anti-cancer drugs since its overexpression is associated with various types of cancers. In the present study, 2D and 3D atom based QSAR study were accomplished with 6 component PLS factor for 230 pyrido[2,3-d]pyramidine correspondents along with flexible ligand docking in the extra precision mode with the application of core constraints followed by the binding energy determinations. Kernel based partial least square analysis fitting with fingerprints initially created worthy models, among which the one with molprint2D fingerprints generated a noble model with a score value of 0.8322. Atom Based 3D QSAR resulted in an effective model with Regression coefficient (R2 = 0.8372), and Q2 = 0.7381. Docking experiments exposed hydrogen bonding interactions with hinge region residues, salt bridge formation and л-л stacking interaction as the leading non-covalent interactions causative of the inhibitory activity of CDK4 inhibitors. The primary factors that induced the stability of protein-ligand complex are the van der Waals interactions, lipophilic interactions and coulombic interactions.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad Cuantitativa , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología
12.
Oncogene ; 37(13): 1685-1698, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29335521

RESUMEN

There is growing interest in studying the molecular mechanisms of crosstalk between cancer metabolism and the cell cycle. 6-phosphate fructose-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) is a well-known glycolytic activator that plays an important role in tumorigenesis. We investigated whether PFKFB3 was directly involved in oncogenic signaling networks. Mass Spectrometry showed that PFKFB3 interacts with cyclin-dependent kinase (CDK) 4, which controls the transition from G1 phase to S phase of the cell cycle. Further analysis indicated that lysine 147 was a key site for the binding of PFKBFB3 to CDK4. PFKFB3 binding resulted in the accumulation of CDK4 protein by inhibiting ubiquitin proteasome degradation mediated by the heat shock protein 90-Cdc37-CDK4 complex. The proteasome-dependent degradation of CDK4 was accelerated by disrupting the interaction of PFKFB3 with CDK4 by mutating lysine (147) to alanine. Blocking PFKFB3-CDK4 interaction improved the therapeutic effect of FDA-approved CDK4 inhibitor palbociclib on breast cancer. These findings suggest that PFKFB3 is a hub for coordinating cell cycle and glucose metabolism. Combined targeting of PFKFB3 and CDK4 may be new strategy for breast cancer treatment.


Asunto(s)
Ciclo Celular , Quinasa 4 Dependiente de la Ciclina/metabolismo , Fosfofructoquinasa-2/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclo Celular/genética , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/genética , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones SCID , Persona de Mediana Edad , Fosfofructoquinasa-2/metabolismo , Piperazinas/uso terapéutico , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Proteolisis/efectos de los fármacos , Piridinas/uso terapéutico
13.
Cancer Sci ; 109(3): 656-665, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29288563

RESUMEN

Cell division cycle 37 (Cdc37) is an important partner for heat shock protein 90 (HSP90), assisting in molecular chaperone activities, particularly with regard to the regulation of protein kinases. Given its influence on cell growth pathways, Cdc37 has been discussed as a potential intermediate in carcinogenesis. However, to date, the potential functional roles and molecular mechanisms by which Cdc37 regulates cell survival in colorectal carcinoma (CRC) remain unclear. Here, we investigated the expression of Cdc37 and its clinical significance in CRC, and systematically explored the role and the underlying mechanism of Cdc37 in CRC cell survival both in vitro and in vivo. Our results showed that Cdc37 was remarkably up-regulated in CRC, which facilitated cell survival mainly by promoting cell proliferation, G1-S transition, and inhibiting cell apoptosis. Our data further indicated that Cdc37 increased the stability of cyclin-dependent kinase 4 (CDK4) to activate the retinoblastoma 1 (RB1) signaling pathway, followed by increased expression of Bcl-2 and Bcl-xL, which ultimately promoted cell survival in CRC. Moreover, knockdown of CDK4 reversed the Cdc37-mediated effect in promoting the progression of CRC. Our findings showed that Cdc37 played a critical role in promoting CRC cell survival by increasing CDK4 stability to activate the RB1 signaling pathway. Thereby, Cdc37 might serve as a potential therapeutic target in CRC patients.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Chaperoninas/genética , Chaperoninas/metabolismo , Neoplasias Colorrectales/patología , Quinasa 4 Dependiente de la Ciclina/metabolismo , Proteínas de Unión a Retinoblastoma/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Quinasa 4 Dependiente de la Ciclina/química , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Trasplante de Neoplasias , Fosforilación , Estabilidad Proteica , Transducción de Señal , Regulación hacia Arriba
15.
Comput Biol Chem ; 71: 224-229, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29153893

RESUMEN

The discovery of ATP competitive CDK4 inhibitors is an on-going challenging task in cancer therapy. Here, an attempt has been made to develop new leads targeting ATP binding site of CDK4 by applying 3D-QSAR pharmacophore mapping and molecular docking methods The outcome of 6 leads offers a significant contribution for selective CDK4 inhibition, since they show potential binding interactions with Val96, Arg101, and Glu144 residues of CDK4, that are unique and from other kinases. It is worth noting that there is a striking similarity in binding interactions of the leads and known CDK4 inhibitors, namely Abemaciclib, Palbociclib and Ribociclib. Further key features, including high dock score value, good predicted activity, scaffold diversity, and the acceptable ADME profile of leads, provide a great opportunity for the development of highly potent and selective ATP competitive inhibitors of CDK4.


Asunto(s)
Adenosina Trifosfato/química , Quinasa 4 Dependiente de la Ciclina/química , Descubrimiento de Drogas , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad Cuantitativa , Aminopiridinas/química , Aminopiridinas/farmacología , Bencimidazoles/química , Bencimidazoles/farmacología , Sitios de Unión/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Humanos , Piperazinas/química , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Purinas/química , Purinas/farmacología , Piridinas/química , Piridinas/farmacología
16.
Mol Biosyst ; 13(11): 2235-2253, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-28926061

RESUMEN

The overarching goal of delineating molecular principles underlying differentiation of the activation mechanisms in cyclin-dependent kinases (CDKs) is important for understanding regulatory divergences among closely related kinases which can be exploited in drug discovery of targeted and allosteric inhibitors. To systematically characterize dynamic, energetic and network signatures of the activation mechanisms, we combined atomistic simulations and elastic network modeling with the analysis of the residue interaction networks and rigidity decomposition of the CDK2-cyclin A and CDK4-cyclin D1/D3 complexes. The results of this study show that divergences in the activation mechanisms of CDK2 and CDK4 may be determined by differences in stabilization and allosteric cooperativity of the regulatory regions. We show that differential stabilization of the kinase lobes in the CDK4-cyclin D complexes caused by the elevated mobility of the N-lobe residues can weaken allosteric interactions between regulatory regions and compromise cooperativity of the inter-lobe motions that is required to trigger activating transitions. Network modelling and percolation analysis were used to emulate thermal unfolding and perform decomposition of rigid and flexible regions in the CDK2 and CDK4 complexes. These simulations showed that the percolation phase transition in the CDK2-cyclin A complexes is highly cooperative and driven by allosteric coupling between functional regions from both kinase lobes. In contrast, the imbalances in the distribution of rigid and flexible regions for the CDK4-cyclin D complexes, which are manifested by the intrinsic instability of the N-lobe, may weaken allosteric interactions and preclude productive activation. The results of this integrative computational study offer a simple and robust network-based model that explains regulatory divergences between CDK2 and CDK4 kinases.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/química , Modelos Moleculares , Conformación Molecular , Algoritmos , Aminoácidos/química , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Relación Estructura-Actividad
17.
EMBO Mol Med ; 9(8): 1052-1066, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28566333

RESUMEN

Cyclin D-CDK4/6 are the first CDK complexes to be activated in the G1 phase in response to oncogenic pathways. The specific CDK4/6 inhibitor PD0332991 (palbociclib) was recently approved by the FDA and EMA for treatment of advanced ER-positive breast tumors. Unfortunately, no reliable predictive tools are available for identifying potentially responsive or insensitive tumors. We had shown that the activating T172 phosphorylation of CDK4 is the central rate-limiting event that initiates the cell cycle decision and signals the presence of active CDK4. Here, we report that the profile of post-translational modification including T172 phosphorylation of CDK4 differs among breast tumors and associates with their subtypes and risk. A gene expression signature faithfully predicted CDK4 modification profiles in tumors and cell lines. Moreover, in breast cancer cell lines, the CDK4 T172 phosphorylation best correlated with sensitivity to PD0332991. This gene expression signature identifies tumors that are unlikely to respond to CDK4/6 inhibitors and could help to select a subset of patients with HER2-positive and basal-like tumors for clinical studies on this class of drugs.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/química , Piperazinas/farmacología , Procesamiento Proteico-Postraduccional , Piridinas/farmacología , Transcriptoma , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Análisis por Micromatrices , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología
18.
J Biol Chem ; 292(24): 10131-10141, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28446612

RESUMEN

CDK4 regulates G1/S phase transition in the mammalian cell cycle by phosphorylating retinoblastoma family proteins. However, the mechanism underlying the regulation of CDK4 activity is not fully understood. Here, we show that CDK4 protein is degraded by anaphase-promoting complex/cyclosome (APC/C) during metaphase-anaphase transition in HeLa cells, whereas its main regulator, cyclin D1, remains intact but is sequestered in cytoplasm. CDK4 protein reaccumulates in the following G1 phase and shuttles between the nucleus and the cytoplasm to facilitate the nuclear import of cyclin D1. Without CDK4, cyclin D1 cannot enter the nucleus. Point mutations that disrupt CDK4 and cyclin D1 interaction impair the nuclear import of cyclin D1 and the activity of CDK4. RNAi knockdown of CDK4 also induces cytoplasmic retention of cyclin D1 and G0/G1 phase arrest of the cells. Collectively, our data demonstrate that CDK4 protein is degraded in late mitosis and reaccumulates in the following G1 phase to facilitate the nuclear import of cyclin D1 for activation of CKD4 to initiate a new cell cycle in HeLa cells.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Fase G1 , Mitosis , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Ciclina D1/química , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/genética , Inducción Enzimática , Estabilidad de Enzimas , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ratones , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Señales de Localización Nuclear/química , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Mutación Puntual , Estabilidad Proteica , Transporte de Proteínas , Proteolisis , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
19.
Science ; 352(6293): 1542-7, 2016 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-27339980

RESUMEN

The Hsp90 molecular chaperone and its Cdc37 cochaperone help stabilize and activate more than half of the human kinome. However, both the mechanism by which these chaperones assist their "client" kinases and the reason why some kinases are addicted to Hsp90 while closely related family members are independent are unknown. Our structural understanding of these interactions is lacking, as no full-length structures of human Hsp90, Cdc37, or either of these proteins with a kinase have been elucidated. Here we report a 3.9 angstrom cryo-electron microscopy structure of the Hsp90-Cdc37-Cdk4 kinase complex. Surprisingly, the two lobes of Cdk4 are completely separated with the ß4-ß5 sheet unfolded. Cdc37 mimics part of the kinase N lobe, stabilizing an open kinase conformation by wedging itself between the two lobes. Finally, Hsp90 clamps around the unfolded kinase ß5 strand and interacts with exposed N- and C-lobe interfaces, protecting the kinase in a trapped unfolded state. On the basis of this structure and an extensive amount of previously collected data, we propose unifying conceptual and mechanistic models of chaperone-kinase interactions.


Asunto(s)
Proteínas de Ciclo Celular/química , Chaperoninas/química , Quinasa 4 Dependiente de la Ciclina/química , Proteínas HSP90 de Choque Térmico/química , Complejos Multiproteicos/química , Desplegamiento Proteico , Animales , Proteínas de Ciclo Celular/ultraestructura , Chaperoninas/ultraestructura , Quinasa 4 Dependiente de la Ciclina/ultraestructura , Estabilidad de Enzimas , Proteínas HSP90 de Choque Térmico/ultraestructura , Humanos , Modelos Moleculares , Complejos Multiproteicos/ultraestructura , Estructura Secundaria de Proteína , Células Sf9
20.
Chembiochem ; 17(8): 737-44, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26946188

RESUMEN

Understanding the intricate steps of protein kinase regulation requires characterization of protein-protein interactions between the catalytic subunit, its regulatory partners and the substrate. Fluorescent probes are useful tools with which to study such interactions and to gain insight into their affinities and specificities. Solvatochromic probes, which display changes in their fluorescence emission in response to changes in the polarity of the medium, are particularly attractive. Here we describe conjugation of a switchable fluorescent dye, TP-2Rho, to peptide and protein derivatives of cyclin-dependent kinase 4 (CDK4) and its application to characterization of the interactions between the catalytic subunit of this kinase, its regulatory partner cyclin D1 and a peptide substrate. We demonstrate the sensitivity of TP-2Rho in relation to of those other dyes used for monitoring peptide-protein and protein-protein interactions. Moreover, we show that TP-Rho-labelled peptides can be introduced into living cells to probe endogenous CDK4/cyclin D.


Asunto(s)
Ciclina D/química , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/metabolismo , Colorantes Fluorescentes/química , Maleimidas/química , Tiazolidinas/química , Colorantes Fluorescentes/síntesis química , Células HeLa , Humanos , Maleimidas/síntesis química , Modelos Moleculares , Estructura Molecular , Unión Proteica , Tiazolidinas/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...