Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Bull Math Biol ; 79(10): 2356-2393, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28852950

RESUMEN

This paper investigates cell proliferation dynamics in small tumor cell aggregates using an individual-based model (IBM). The simulation model is designed to study the morphology of the cell population and of the cell lineages as well as the impact of the orientation of the division plane on this morphology. Our IBM model is based on the hypothesis that cells are incompressible objects that grow in size and divide once a threshold size is reached, and that newly born cell adhere to the existing cell cluster. We performed comparisons between the simulation model and experimental data by using several statistical indicators. The results suggest that the emergence of particular morphologies can be explained by simple mechanical interactions.


Asunto(s)
Linaje de la Célula , Modelos Biológicos , Neoplasias/patología , Algoritmos , Fenómenos Biomecánicos , División Celular , Línea Celular Tumoral , Linaje de la Célula/fisiología , Proliferación Celular , Tamaño de la Célula , Simulación por Computador , Células HCT116 , Humanos , Conceptos Matemáticos , Microscopía por Video , Neoplasias/fisiopatología
2.
BMC Cancer ; 13: 73, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23394599

RESUMEN

BACKGROUND: MultiCellular Tumor Spheroid (MCTS) mimics the organization of a tumor and is considered as an invaluable model to study cancer cell biology and to evaluate new antiproliferative drugs. Here we report how the characteristics of MCTS in association with new technological developments can be used to explore the regionalization and the activation of cell cycle checkpoints in 3D. METHODS: Cell cycle and proliferation parameters were investigated in Capan-2 spheroids by immunofluorescence staining, EdU incorporation and using cells engineered to express Fucci-red and -green reporters. RESULTS: We describe in details the changes in proliferation and cell cycle parameters during spheroid growth and regionalization. We report the kinetics and regionalized aspects of cell cycle arrest in response to checkpoint activation induced by EGF starvation, lovastatin treatment and etoposide-induced DNA damage. CONCLUSION: Our data present the power and the limitation of spheroids made of genetically modified cells to explore cell cycle checkpoints. This study paves the way for the investigation of molecular aspects and dynamic studies of the response to novel antiproliferative agents in 3D models.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias Pancreáticas/patología , Esferoides Celulares/patología , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Citotoxinas/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Modelos Biológicos , Neoplasias Pancreáticas/tratamiento farmacológico , Esferoides Celulares/efectos de los fármacos , Células Tumorales Cultivadas , Gemcitabina
3.
PLoS One ; 14(5): e0217227, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31120960

RESUMEN

Growing multicellular spheroids recapitulate many features of expanding microtumours, and therefore they are an attractive system for biomechanical studies. Here, we report an original approach to measure and characterize the forces exerted by proliferating multicellular spheroids. As force sensors, we used high aspect ratio PDMS pillars arranged as a ring that supports a growing breast tumour cell spheroid. After optical imaging and determination of the force application zones, we combined 3D reconstruction of the shape of each deformed PDMS pillar with the finite element method to extract the forces responsible for the experimental observation. We found that the force exerted by growing spheroids ranges between 100nN and 300nN. Moreover, the exerted force was dependent on the pillar stiffness and increased over time with spheroid growth.


Asunto(s)
Neoplasias de la Mama/patología , Técnicas de Cultivo de Célula/instrumentación , Técnicas Analíticas Microfluídicas/instrumentación , Esferoides Celulares/patología , Femenino , Humanos , Estrés Mecánico , Análisis de Matrices Tisulares
4.
Mol Cancer Ther ; 6(1): 318-25, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17237290

RESUMEN

The CDC25 cell cycle regulators are promising targets for new pharmacologic approaches in cancer therapy. Inhibitory compounds such as BN82685 have proven to be effective in specifically targeting CDC25 in cultured cells and in inhibiting tumor cell growth. Here, we report that BN82685 impairs microtubule dynamic instability and alters microtubule organization and assembly at the centrosome in interphase cells. Treatment of mitotic cells with BN82685 delays mitotic spindle assembly, chromosome capture, and metaphase plate formation. Furthermore, we show that combining low concentrations of both BN82685 and paclitaxel inhibits the proliferation of HT29 human colon cancer cells. Our results show a role for CDC25 phosphatases in regulating microtubule dynamics throughout the cell cycle and suggest that combinations of CDC25 inhibitors with microtubule-targeting agents may be of therapeutic value.


Asunto(s)
Benzoquinonas/farmacología , Inhibidores Enzimáticos/farmacología , Interfase/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Tiazoles/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Cromosomas Humanos/efectos de los fármacos , Sinergismo Farmacológico , Células HT29 , Células HeLa , Humanos , Metafase/efectos de los fármacos , Prometafase/efectos de los fármacos
5.
Sci Rep ; 7: 44939, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28322312

RESUMEN

Tissue mimics (TMs) on the scale of several hundred microns provide a beneficial cell culture configuration for in vitro engineered tissue and are currently under the spotlight in tissue engineering and regenerative medicine. Due to the cell density and size, TMs are fairly inaccessible to optical observation and imaging within these samples remains challenging. Light Sheet Fluorescence Microscopy (LSFM)- an emerging and attractive technique for 3D optical sectioning of large samples- appears to be a particularly well-suited approach to deal with them. In this work, we compared the effectiveness of different light sheet illumination modalities reported in the literature to improve resolution and/or light exposure for complex 3D samples. In order to provide an acute and fair comparative assessment, we also developed a systematic, computerized benchmarking method. The outcomes of our experiment provide meaningful information for valid comparisons and arises the main differences between the modalities when imaging different types of TMs.


Asunto(s)
Biomimética/métodos , Imagenología Tridimensional/métodos , Microscopía Fluorescente/métodos , Imagen Óptica/métodos , Animales , Humanos , Miocitos Cardíacos/metabolismo , Ratas , Imagen de Lapso de Tiempo
6.
Cancer Res ; 64(9): 3320-5, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15126376

RESUMEN

CDC25 dual-specificity phosphatases are essential regulators that dephosphorylate and activate cyclin-dependent kinase/cyclin complexes at key transitions of the cell cycle. CDC25 activity is currently considered to be an interesting target for the development of new antiproliferative agents. Here we report the identification of a new CDC25 inhibitor and the characterization of its effects at the molecular and cellular levels, and in animal models. BN82002 inhibits the phosphatase activity of recombinant human CDC25A, B, and C in vitro. It impairs the proliferation of tumoral cell lines and increases cyclin-dependent kinase 1 inhibitory tyrosine phosphorylation. In synchronized HeLa cells, BN82002 delays cell cycle progression at G1-S, in S phase and at the G2-M transition. In contrast, BN82002 arrests U2OS cell cycle mostly in the G1 phase. Selectivity of this inhibitor is demonstrated: (a) by the reversion of the mitotic-inducing effect observed in HeLa cells upon CDC25B overexpression; and (b) by the partial reversion of cell cycle arrest in U2OS expressing CDC25. We also show that BN82002 reduces growth rate of human tumor xenografts in athymic nude mice. BN82002 is a original CDC25 inhibitor that is active both in cell and animal models. This greatly reinforces the interest in CDC25 as an anticancer target.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Etilaminas , Femenino , Células HeLa , Humanos , Ratones , Ratones Desnudos , Mitosis/efectos de los fármacos , Nitrocompuestos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/biosíntesis , Fosfatasas cdc25/genética
7.
Oncogene ; 22(2): 220-32, 2003 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-12527891

RESUMEN

Human dual-specificity phosphatases CDC25 (A, B and C) play an important role in the control of cell cycle progression by activating the cyclin-dependent kinases (CDKs). Regulation of these phosphatases during the cell cycle involves post-translational modifications such as phosphorylation and protein-protein interactions. Given the suspected involvement of the protein kinase CK2 at the G2/M transition, we have investigated its effects on the CDC25B phosphatase. We show that in vitro CK2 phosphorylates CDC25B, but not CDC25C. Mass spectrometry analysis demonstrates that at least two serine residues, Ser-186 and Ser-187, are phosphorylated in vivo. We also report that CDC25B interacts with CK2, and this interaction, mediated by the CK2beta regulatory subunit, involves domains that are located within the first 55 amino acids of CK2beta and between amino acids 122 and 200 on CDC25B. This association was confirmed in vivo, in Sf9 insect cells and in U(2)OS human cells expressing an HA epitope-tagged CDC25B. Finally, we demonstrate that phosphorylation of CDC25B by protein kinase CK2 increases the catalytic activity of the phosphatase in vitro as well as in vivo. We discuss the possibility that CDC25B phosphorylation by CK2 could play a role in the regulation of the activity of CDC25B as a starter of mitosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fosfatasas cdc25/metabolismo , Secuencia de Aminoácidos , Animales , Quinasa de la Caseína II , Proteínas de Ciclo Celular/genética , Células Cultivadas , Epítopos/genética , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/metabolismo , Spodoptera/citología , Regulación hacia Arriba , Fosfatasas cdc25/genética
8.
Cancer Res ; 75(12): 2426-33, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25855380

RESUMEN

Cell aggregation is frequently impaired during the growth of primary tumors and the formation of metastatic lesions. Cell aggregation depends on cell-cell adhesion; however, no rigorous approach exists to monitor and quantify it accurately in the absence of the confounding factors of cell-substrate adhesion and the resulting cell motility on the substrate. We report here a highly reproducible, automated, microscopy-based quantification of tumor-cell spheroid formation in the absence of cell-substrate adhesion and use it to characterize cell aggregation dynamics in the early steps of this process. This method is based on fluorescence and bright-field microscopy and on a custom MATLAB program to quantify automatically the cells' aggregation kinetics. We demonstrate that the cell-cell adhesion protein E-cadherin and the desmosome proteins DSG2 and DSC2 are important for aggregation. Furthermore, we show that inhibition or silencing of myosin IIa enhances aggregation, suggesting that cytoskeleton tension inhibits tumor cell aggregation. This work opens new avenues to study the principles that govern multicellular aggregation, to characterize the aggregation properties of various tumor cell types, as well as to screen for drugs that inhibit or promote aggregation.


Asunto(s)
Adhesión Celular/fisiología , Agregación Celular/fisiología , Comunicación Celular/fisiología , Neoplasias/patología , Cadherinas/metabolismo , Movimiento Celular/fisiología , Citoesqueleto/patología , Desmocolinas/metabolismo , Desmogleína 2/metabolismo , Células HCT116 , Humanos , Neoplasias/metabolismo , Transfección
9.
Cancer Res ; 71(5): 1968-77, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21363925

RESUMEN

CDC25B phosphatases must activate cyclin B-CDK1 complexes to restart the cell cycle after an arrest in G2 phase caused by DNA damage. However, little is known about the precise mechanisms involved in this process, which may exert considerable impact on cancer susceptibility and therapeutic responses. Here we report the discovery of novel N-terminally truncated CDC25B isoforms, referred to as ΔN-CDC25B, with an exclusively nuclear and nonredundant function in cell cycle re-initiation after DNA damage. ΔN-CDC25B isoforms are expressed from a distinct promoter not involved in expression of canonical full-length isoforms. Remarkably, in contrast to the high lability and spatial dynamism of the full-length isoforms, ΔN-CDC25B isoforms are highly stable and exclusively nuclear, strongly suggesting the existence of two pools of CDC25B phosphatases in the cell that have functionally distinct properties. Using isoform-specific siRNA, we found that depleting full-length isoforms, but not ΔN-CDC25B isoforms, delays entry into mitosis. Thus, in an unperturbed cell cycle, the full-length isoforms are exclusively responsible for activating cyclin B-CDK1. Strikingly, in the late response to DNA damage, we found a CHK1-dependent shift in accumulation of CDC25B isoforms toward the ΔN-CDC25B species. Under this physiological stress condition, the ΔN-CDC25B isoform was found to play a crucial, nonredundant function in restarting the cell cycle after DNA damage-induced G2 phase arrest. Our findings reveal the existence of a previously unrecognized CDC25B isoform that operates specifically in the nucleus to reinitiate G2/M transition after DNA damage.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , División Celular/efectos de los fármacos , Fase G2/genética , Fosfatasas cdc25/metabolismo , Western Blotting , Línea Celular Tumoral , Núcleo Celular/metabolismo , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño , Fosfatasas cdc25/genética
10.
Cell Cycle ; 9(18): 3815-22, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20930503

RESUMEN

Tight regulation of cell cycle progression is essential for the maintenance of genomic integrity in response to DNA injury. The aim of this study was to identify new deubiquitinating enzymes (DUBs) involved in the regulation of the G2/M checkpoint. By using an siRNA-based screen to identify DUBs with an inherent ability to enhance a CDC25B-dependent G2/M checkpoint bypass, we have identified 11 candidates whose invalidation compromises checkpoint stringency. We subsequently focused our attention on one of these, the previously uncharacterized USP50. Using a TAP-tag approach associated to mass spectrometry, in addition to a yeast-two-hybrid screen, we identified HSP90 as a major interacting partner for USP50. We also demonstrate USP50 depletion causes a loss in accumulation of the HSP90 client Wee1, which is an essential component of the G2/M cell cycle arrest. Finally, we show that in response to DNA damaging agents, USP50 accumulates in the nucleus. We propose that USP50 may act through a HSP90-dependent mechanism to counteract CDC25B mitotic inducing activity and prevent Wee1 degradation, thereby repressing entry into mitosis following activation of the DNA damage checkpoint.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Endopeptidasas/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/metabolismo , División Celular , Línea Celular , Núcleo Celular/metabolismo , Daño del ADN , Endopeptidasas/genética , Fase G2 , Humanos , Espectrometría de Masas , Estabilidad Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Técnicas del Sistema de Dos Híbridos , Proteasas Ubiquitina-Específicas , Fosfatasas cdc25/metabolismo
11.
Cell Cycle ; 4(9): 1233-8, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16082213

RESUMEN

CDC25B is one of the three human dual-specificity phosphatases involved in the activation of cyclin-dependent kinases at key stages of the cell division cycle. CDC25B that is responsible for the activation of CDK1-cyclin B1 is regulated by phosphorylation. The STK15/Aurora-A kinase locally phosphorylates CDC25B on serine 353 at the centrosome during the G2/M transition. Here we have investigated this phosphorylation event during the cell cycle, and in response to activation of the G2 DNA damage checkpoint. We show that accumulation of the S353-phosphorylated form of CDC25B at the centrosome correlates with the relocalization of cyclin B1 to the nucleus and the activation of CDK1 at entry into mitosis. Upon activation of the G2/M checkpoint by DNA damage, we demonstrate that Aurora-A is not activated and consequently CDC25B is not phosphorylated. We show that ectopic expression of Aurora-A results in a bypass of the checkpoint that was partially overcome by a S353A mutant of CDC25B. Finally, we show that bypass of the G2/M checkpoint by the CHK1 kinase inhibitor UCN-01 results in the activation of Aurora-A and phosphorylation of CDC25B on S353. These results strongly suggest that Aurora-A-mediated phosphorylation of CDC25B at the centrosome is an important step contributing to the earliest events inducing mitosis, upstream of CDK1-cyclin B1 activation.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Daño del ADN , Proteínas Serina-Treonina Quinasas/química , Fosfatasas cdc25/fisiología , Aurora Quinasa A , Aurora Quinasas , Proteínas de Ciclo Celular/metabolismo , División Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Centrosoma/metabolismo , Ciclina B/química , Ciclina B1 , Fase G2 , Células HeLa , Histonas/química , Humanos , Microscopía Fluorescente , Mitosis , Mutación , Fosforilación , Conformación Proteica , Serina/química , Factores de Tiempo , Transfección , Tirosina/química , Fosfatasas cdc25/metabolismo
12.
J Biol Chem ; 277(38): 35176-82, 2002 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-12107172

RESUMEN

CDC25B phosphatases are essential regulators that control cyclin-dependent kinases activities at the entry into mitosis. In this study, we demonstrate that serine 146 is required for two crucial features of CDC25B1. It is essential for CDC25B1 to function as a mitotic inducer and to prevent CDC25B1 export from the nucleus. We also show that serine 146 is phosphorylated in vitro by CDK1-cyclin B. However, phosphorylation of CDC25B does not stimulate its phosphatase activity, and mutation of serine 146 had no effect on its catalytic activity. Serine 146 phosphorylation is proposed to be a key event in the regulation of the CDC25B function in the initiation of mammalian mitosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Mitosis/fisiología , Serina/metabolismo , Fosfatasas cdc25/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiología , Humanos , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas , Fosfatasas cdc25/química , Fosfatasas cdc25/fisiología
13.
Biol Cell ; 95(8): 547-54, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14630392

RESUMEN

Regulation of the intracellular localisation of its actors is one of the key mechanisms underlying cell cycle control. CDC25 phosphatases are activators of Cyclin-Dependent Kinases (CDK) that undergo nucleo-cytoplasmic shuttling during the cell cycle and in response to checkpoint activation. Here we report that the protein kinase PKB/Akt phosphorylates CDC25B on serine 353, resulting in a nuclear export-dependent cytoplasmic accumulation of the phosphatase. Oxidative stress activates PKB/Akt and reproduces the effect on CDC25B phosphorylation and localisation. However, inhibition of PKB/Akt activity only partially reverted the effect of oxidative stress on CDC25B localisation and mutation of serine 353 abolishes phosphorylation but only delays nuclear exclusion. These results indicate that additional mechanisms are also involved in preventing nuclear import of CDC25B. Our findings identify CDC25B as a target of PKB/Akt and provide new insight into the regulation of its localisation in response to stress-activated signalling pathways.


Asunto(s)
Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Citoplasmáticos y Nucleares , Fosfatasas cdc25/análisis , Fosfatasas cdc25/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/química , Vectores Genéticos , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Carioferinas/fisiología , Señales de Localización Nuclear/fisiología , Estrés Oxidativo , Plásmidos , Mutación Puntual , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes/genética , Fosfatasas cdc25/genética , Proteína Exportina 1
14.
J Cell Sci ; 117(Pt 12): 2523-31, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15128871

RESUMEN

Aurora-A protein kinase, which is the product of an oncogene, is required for the assembly of a functional mitotic apparatus and the regulation of cell ploidy. Overexpression of Aurora-A in tumour cells has been correlated with cancer susceptibility and poor prognosis. Aurora-A activity is required for the recruitment of CDK1-cyclin B1 to the centrosome prior to its activation and the commitment of the cell to mitosis. In this report, we demonstrate that the CDC25B phosphatase, an activator of cyclin dependent kinases at mitosis, is phosphorylated both in vitro and in vivo by Aurora-A on serine 353 and that this phosphorylated form of CDC25B is located at the centrosome during mitosis. Knockdown experiments by RNAi confirm that the centrosome phosphorylation of CDC25B on S353 depends on Aurora-A kinase. Microinjection of antibodies against phosphorylated S353 results in a mitotic delay whilst overexpression of a S353 phosphomimetic mutant enhances the mitotic inducing effect of CDC25B. Our results demonstrate that Aurora-A phosphorylates CDC25B in vivo at the centrosome during mitosis. This phosphorylation might locally participate in the control of the onset of mitosis. These findings re-emphasise the role of the centrosome as a functional integrator of the pathways contributing to the triggering of mitosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Centrosoma/metabolismo , Fase G2/fisiología , Proteínas Quinasas/metabolismo , Fosfatasas cdc25/metabolismo , Anticuerpos/metabolismo , Anticuerpos Monoclonales/metabolismo , Aurora Quinasas , Proteínas de Ciclo Celular/química , Células HeLa , Humanos , Microinyecciones , Fosforilación , Proteínas Serina-Treonina Quinasas , Interferencia de ARN , Serina/metabolismo , Factores de Tiempo , Proteínas de Xenopus , Fosfatasas cdc25/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA