Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Cell Physiol ; 232(9): 2461-2468, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27608139

RESUMEN

In live cell imaging of fluorescent proteins, phototoxicity of the excitation light can be problematical. Cell death is obvious, but reduced cell viability can make the interpretation of observations error prone. We characterized the phototoxic consequences of 488 and 546 nm light on untransformed human cells and tested methods that have or could be used to alleviate photodamage. Unlabeled RPE1 cells were given single 0.5-2.5 min irradiations in early G1 from a mercury arc lamp on a fluorescence microscope. Four hundred eighty-eight nanometer light produced a dose-dependent decrease in the percentage of cells that progressed to mitosis, slowing of the cell cycle for some of those entering mitosis, and a ∼12% incidence of cell death for the highest dose. For 546 nm light we found a 10-15% reduction in the percentage of cells entering mitosis, no strong dose dependency, and a ∼2% incidence of cell death for the longest irradiations. For cells expressing GFP-centrin1 or mCherry-centrin1, fewer entered mitosis for each dose than unlabeled cells. For constant total dose 488 nm light irradiations of unlabeled cells, reducing the intensity 10-fold or spreading the exposures out as a series of 10 sec pulses at 1 min intervals produced a minor and not consistent improvement in the percentage of cells entering mitosis. Reducing oxidative processes, by culturing at ∼3% oxygen or adding the reducing agent Trolox noticeably increased the fraction of cells entering mitosis. Thus, for long-term imaging there can be value to using RFP constructs and for GFP-tagged proteins reducing oxidative processes. J. Cell. Physiol. 232: 2461-2468, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de la radiación , Células Epiteliales/efectos de la radiación , Luz/efectos adversos , Microscopía Fluorescente/efectos adversos , Mitosis/efectos de la radiación , Epitelio Pigmentado de la Retina/efectos de la radiación , Antioxidantes/farmacología , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Línea Celular , Cromanos/farmacología , Citoprotección , Relación Dosis-Respuesta en la Radiación , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Mitosis/efectos de los fármacos , Estrés Oxidativo , Oxígeno/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Factores de Tiempo , Transfección , Proteína Fluorescente Roja
2.
J Cell Physiol ; 230(11): 2683-94, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25808524

RESUMEN

The Brahma (BRM) and Brahma-related Gene 1 (BRG1) ATPases are highly conserved homologs that catalyze the chromatin remodeling functions of the multi-subunit human SWI/SNF chromatin remodeling enzymes in a mutually exclusive manner. SWI/SNF enzyme subunits are mutated or missing in many cancer types, but are overexpressed without apparent mutation in other cancers. Here, we report that both BRG1 and BRM are overexpressed in most primary breast cancers independent of the tumor's receptor status. Knockdown of either ATPase in a triple negative breast cancer cell line reduced tumor formation in vivo and cell proliferation in vitro. Fewer cells in S phase and an extended cell cycle progression time were observed without any indication of apoptosis, senescence, or alterations in migration or attachment properties. Combined knockdown of BRM and BRG1 showed additive effects in the reduction of cell proliferation and time required for completion of cell cycle, suggesting that these enzymes promote cell cycle progression through independent mechanisms. Knockout of BRG1 or BRM using CRISPR/Cas9 technology resulted in the loss of viability, consistent with a requirement for both enzymes in triple negative breast cancer cells.


Asunto(s)
Proliferación Celular/genética , ADN Helicasas/biosíntesis , Proteínas Nucleares/biosíntesis , Factores de Transcripción/biosíntesis , Neoplasias de la Mama Triple Negativas/genética , Adenosina Trifosfatasas/biosíntesis , Adenosina Trifosfatasas/genética , Animales , Sistemas CRISPR-Cas , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Proteínas Nucleares/genética , Factores de Transcripción/genética , Neoplasias de la Mama Triple Negativas/patología
3.
J Cell Physiol ; 229(10): 1427-36, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24532022

RESUMEN

The radiation and radiomimetic drugs used to treat human tumors damage DNA in both cancer cells and normal proliferating cells. Centrosome amplification after DNA damage is well established for transformed cell types but is sparsely reported and not fully understood in untransformed cells. We characterize centriole behavior after DNA damage in synchronized untransformed human cells. One hour treatment of S phase cells with the radiomimetic drug, Doxorubicin, prolongs G2 by at least 72 h, though 14% of the cells eventually go through mitosis in that time. By 72 h after DNA damage we observe a 52% incidence of centriole disengagement plus a 10% incidence of extra centrioles. We find that either APC/C or Plk activities can disengage centrioles after DNA damage, though they normally work in concert. All disengaged centrioles are associated with γ-tubulin and maturation markers and thus, should in principle be capable of reduplicating and organizing spindle poles. The low incidence of reduplication of disengaged centrioles during G2 is due to the p53-dependent expression of p21 and the consequent loss of Cdk2 activity. We find that 26% of the cells going through mitosis after DNA damage contain disengaged or extra centrioles. This could produce genomic instability through transient or persistent spindle multipolarity. Thus, for cancer patients the use of DNA damaging therapies raises the chances of genomic instability and evolution of transformed characteristics in proliferating normal cell populations.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/inducido químicamente , Centriolos/efectos de los fármacos , Daño del ADN , Doxorrubicina/toxicidad , Epitelio Pigmentado de la Retina/efectos de los fármacos , Ciclosoma-Complejo Promotor de la Anafase/antagonistas & inhibidores , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Centriolos/metabolismo , Centriolos/patología , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Inestabilidad Genómica , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Factores de Tiempo , Transfección , Tubulina (Proteína)/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Quinasa Tipo Polo 1
4.
J Cell Biol ; 176(2): 173-82, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17227892

RESUMEN

How centrosome removal or perturbations of centrosomal proteins leads to G1 arrest in untransformed mammalian cells has been a mystery. We use microsurgery and laser ablation to remove the centrosome from two types of normal human cells. First, we find that the cells assemble centrioles de novo after centrosome removal; thus, this phenomenon is not restricted to transformed cells. Second, normal cells can progress through G1 in its entirety without centrioles. Therefore, the centrosome is not a necessary, integral part of the mechanisms that drive the cell cycle through G1 into S phase. Third, we provide evidence that centrosome loss is, functionally, a stress that can act additively with other stresses to arrest cells in G1 in a p38-dependent fashion.


Asunto(s)
Ciclo Celular/fisiología , Centriolos/fisiología , Centrosoma/fisiología , Células Epiteliales/metabolismo , Bromodesoxiuridina/metabolismo , Proteínas de Unión al Calcio/análisis , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Células Cultivadas , Centriolos/química , Centriolos/ultraestructura , Proteínas Cromosómicas no Histona/análisis , Inhibidores Enzimáticos/farmacología , Células Epiteliales/citología , Células Epiteliales/ultraestructura , Fase G1/fisiología , Humanos , Imidazoles/farmacología , Luz , Microscopía Electrónica , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Curr Opin Cell Biol ; 16(1): 49-54, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15037304

RESUMEN

Centrosome amplification (the presence of more than two centrosomes at mitosis) is characteristic of many human cancers. Extra centrosomes can cause the assembly of multipolar spindles, which unequally distribute chromosomes to daughter cells; the resulting genetic imbalances may contribute to cellular transformation. However, this raises the question of how a population of cells with centrosome amplification can survive such chaotic mitoses without soon becoming non-viable as a result of chromosome loss. Recent observations indicate that a variety of mechanisms partially mute the practical consequences of centrosome amplification. Consequently, populations of cells propagate with good efficiency, despite centrosome amplification, yet have an elevated mitotic error rate that can fuel the evolution of the transformed state.


Asunto(s)
Centrosoma/metabolismo , Neoplasias/ultraestructura , Animales , Humanos , Mitosis , Neoplasias/genética , Neoplasias/metabolismo , Huso Acromático/metabolismo
6.
Curr Biol ; 17(23): 2081-6, 2007 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-18060787

RESUMEN

For mammalian somatic cells, the importance of microtubule cytoskeleton integrity during interphase cell-cycle progression is uncertain. The loss, suppression, or stabilization of the microtubule cytoskeleton has been widely reported to cause a G1 arrest in a variable, and often high, proportion of cell populations, suggesting the existence of a "microtubule damage," "microtubule integrity," or "postmitotic" checkpoint in G1 or G2. We found that when normal human cells (hTERT RPE1 and primary fibroblasts) are continuously exposed to nocodazole, they remain in mitosis for 10-48 hr before they slip out of mitosis and arrest in G1; this finding is consistent with previous reports. To eliminate the persistent effects of prolonged mitosis, we isolated anaphase-telophase cells that were just finishing a mitosis of normal duration, then we rapidly and completely disassembled microtubules by chilling the preparations to 0 degrees C for 10 minutes in the continuous presence of nocodazole or colcemid treatment to ensure that the cells entered G1 without a microtubule cytoskeleton. Without microtubules, cells progressed from anaphase to a subsequent mitosis with essentially normal kinetics. Similar results were obtained for cells in which the microtubule cytoskeleton was partially diminished by lower nocodazole doses or augmented and stabilized with taxol. Thus, after a preceding mitosis of normal duration, the integrity of the microtubule cytoskeleton is not subject to checkpoint surveillance, nor is it required for the normal human cell to progress through G1 and the remainder of interphase.


Asunto(s)
Anafase/efectos de los fármacos , Anafase/fisiología , Ciclo Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular , Citoesqueleto/metabolismo , Fibroblastos , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía por Video , Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , Mitosis/fisiología , Nocodazol/farmacología , Paclitaxel/farmacología , Telofase/efectos de los fármacos , Telofase/fisiología
7.
J Virol ; 83(6): 2756-64, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19129456

RESUMEN

High-risk types of human papillomavirus (HPV) are considered the major causative agents of cervical carcinoma. The transforming ability of HPV resides in the E6 and E7 oncogenes, yet the pathway to transformation is not well understood. Cells expressing the oncogene E7 from high-risk HPVs have a high incidence of polyploidy, which has been shown to occur as an early event in cervical carcinogenesis and predisposes the cells to aneuploidy. The mechanism through which E7 contributes to polyploidy is not known. It has been hypothesized that E7 induces polyploidy in response to mitotic stress by abrogating the mitotic spindle assembly checkpoint. It was also proposed that E7 may stimulate rereplication to induce polyploidy. We have tested these hypotheses by using human epithelial cells in which E7 expression induces a significant amount of polyploidy. We find that E7-expressing cells undergo normal mitoses with an intact spindle assembly checkpoint and that they are able to complete cytokinesis. Our results also exclude DNA rereplication as a major mechanism of polyploidization in E7-expressing cells upon microtubule disruption. Instead, we have shown that while normal cells arrest at the postmitotic checkpoint after adaptation to the spindle assembly checkpoint, E7-expressing cells replicate their DNA and propagate as polyploid cells. Thus, abrogation of the postmitotic checkpoint leads to polyploidy formation in E7-expressing human epithelial cells. Our results suggest that downregulation of pRb is important for E7 to induce polyploidy and abrogation of the postmitotic checkpoint.


Asunto(s)
División Celular , Queratinocitos/virología , Papillomaviridae/crecimiento & desarrollo , Proteínas E7 de Papillomavirus/biosíntesis , Poliploidía , Células Cultivadas , Humanos
8.
J Cell Biol ; 168(5): 713-22, 2005 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-15738265

RESUMEN

It has been reported that nontransformed mammalian cells become arrested during G1 in the absence of centrioles (Hinchcliffe, E., F. Miller, M. Cham, A. Khodjakov, and G. Sluder. 2001. Science. 291:1547-1550). Here, we show that removal of resident centrioles (by laser ablation or needle microsurgery) does not impede cell cycle progression in HeLa cells. HeLa cells born without centrosomes, later, assemble a variable number of centrioles de novo. Centriole assembly begins with the formation of small centrin aggregates that appear during the S phase. These, initially amorphous "precentrioles" become morphologically recognizable centrioles before mitosis. De novo-assembled centrioles mature (i.e., gain abilities to organize microtubules and replicate) in the next cell cycle. This maturation is not simply a time-dependent phenomenon, because de novo-formed centrioles do not mature if they are assembled in S phase-arrested cells. By selectively ablating only one centriole at a time, we find that the presence of a single centriole inhibits the assembly of additional centrioles, indicating that centrioles have an activity that suppresses the de novo pathway.


Asunto(s)
Ciclo Celular/fisiología , Centriolos/fisiología , Genes Reporteros , Células HeLa , Humanos , Fase S/fisiología
9.
Cell Biol Int ; 34(12): 1239-45, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21067522

RESUMEN

In preparation for mitosis, the centrosome doubles once and only once to provide the two poles of the mitotic spindle. The presence of more than two centrosomes increases the chances that mitosis will be multipolar, and chromosomes will be distributed unequally. Since the number of mother-daughter centriole pairs determines the number of centrosomes, it is important that only one daughter centriole is assembled at, but slightly separated from, the proximal end of each mother centriole. This numerical and spatial specificity has led to the belief that a 'template' on the mother centriole provides a unique site for procentriole assembly. We review observations that are leading to the demise of this intuitively attractive idea. In its place, we are left with the notion that pericentriolar material at the wall of the mother centriole provides a local environment that promotes the assembly of a macromolecular complex that seeds the daughter centriole. Even though the system normally behaves in a digital fashion to go from zero to just one daughter centriole per mother, this behaviour appears to be based in the precise analogue control of multiple proteins, their activities, and the structure provided by the mother centriole.


Asunto(s)
Centriolos/fisiología , Mitosis/genética , Animales , Fenómenos Fisiológicos Celulares , Centriolos/genética , Centrosoma/fisiología , Humanos , Mitosis/fisiología , Modelos Biológicos , Huso Acromático/genética , Huso Acromático/fisiología
11.
J Cell Biol ; 165(5): 609-15, 2004 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-15184397

RESUMEN

Failure of cells to cleave at the end of mitosis is dangerous to the organism because it immediately produces tetraploidy and centrosome amplification, which is thought to produce genetic imbalances. Using normal human and rat cells, we reexamined the basis for the attractive and increasingly accepted proposal that normal mammalian cells have a "tetraploidy checkpoint" that arrests binucleate cells in G1, thereby preventing their propagation. Using 10 microM cytochalasin to block cleavage, we confirm that most binucleate cells arrest in G1. However, when we use lower concentrations of cytochalasin, we find that binucleate cells undergo DNA synthesis and later proceed through mitosis in >80% of the cases for the hTERT-RPE1 human cell line, primary human fibroblasts, and the REF52 cell line. These observations provide a functional demonstration that the tetraploidy checkpoint does not exist in normal mammalian somatic cells.


Asunto(s)
Fase G1/genética , Genes cdc/fisiología , Mitosis/genética , Poliploidía , Animales , Línea Celular Transformada , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/ultraestructura , Citocalasina D/farmacología , ADN/biosíntesis , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratas
12.
J Cell Biol ; 158(7): 1171-81, 2002 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-12356862

RESUMEN

The centrosome usually replicates in a semiconservative fashion, i.e., new centrioles form in association with preexisting "maternal" centrioles. De novo formation of centrioles has been reported for a few highly specialized cell types but it has not been seen in vertebrate somatic cells. We find that when centrosomes are completely destroyed by laser microsurgery in CHO cells arrested in S phase by hydroxyurea, new centrosomes form by de novo assembly. Formation of new centrosomes occurs in two steps: approximately 5-8 h after ablation, clouds of pericentriolar material (PCM) containing gamma-tubulin and pericentrin appear in the cell. By 24 h, centrioles have formed inside of already well-developed PCM clouds. This de novo pathway leads to the formation of a random number of centrioles (2-14 per cell). Although clouds of PCM consistently form even when microtubules are completely disassembled by nocodazole, the centrioles are not assembled under these conditions.


Asunto(s)
Centrosoma/fisiología , Microtúbulos/metabolismo , Fase S/fisiología , Huso Acromático , Animales , Células CHO/fisiología , Células CHO/ultraestructura , Centrosoma/ultraestructura , Cricetinae , Proteínas Fluorescentes Verdes , Cinética , Rayos Láser , Proteínas Luminiscentes/metabolismo , Microscopía Electrónica , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo
13.
Cancer Res ; 67(6): 2603-10, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363579

RESUMEN

Polyploidy is often an early event during cervical carcinogenesis, and it predisposes cells to aneuploidy, which is thought to play a causal role in tumorigenesis. Cervical and anogenital cancers are induced by the high-risk types of human papillomavirus (HPV). The HPV E6 oncoprotein induces polyploidy in human keratinocytes, yet the mechanism is not known. It was believed that E6 induces polyploidy by abrogating the spindle checkpoint after mitotic stress. We have tested this hypothesis using human keratinocytes in which E6 expression induces a significant amount of polyploidy. We found that E6 expression does not affect the spindle checkpoint. Instead, we provide direct evidence that E6 is capable of abrogating the subsequent G(1) arrest after adaptation of the mitotic stress. E6 targets p53 for degradation, and previous studies have shown an important role for p53 in modulation of the G(1) arrest after mitotic stress. Importantly, we have discovered that E6 mutants defective in p53 degradation also induce polyploidy, although with lower efficiency. These results suggest that E6 is able to induce polyploidy via both p53-dependent and p53-independent mechanisms. Therefore, our studies highlight a novel function of HPV E6 that may contribute to HPV-induced carcinogenesis and improve our understanding of the onset of the disease.


Asunto(s)
Papillomavirus Humano 16/genética , Queratinocitos/fisiología , Mitosis/genética , Proteínas Oncogénicas Virales/genética , Poliploidía , Proteínas Represoras/genética , Proteína p53 Supresora de Tumor/fisiología , Replicación del ADN , Humanos , Queratinocitos/citología , Queratinocitos/virología , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
J Cell Physiol ; 217(3): 626-31, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18651565

RESUMEN

When protein synthesis is completely blocked from before fertilization, the sea urchin zygote arrests in first S phase and the paternal centrosome reduplicates multiple times. However, when protein synthesis is blocked starting in prophase of first mitosis, the zygote divides and the blastomeres arrest in a G1-like state. The centrosome inherited from this mitosis duplicates only once in each blastomere for reasons that are not understood. The late G1 rise in cyclin E/cdk2 kinase activity initiates centrosome duplication in mammalian cells and its activity is needed for centrosome duplication in Xenopus egg extracts. Since the half-time for cyclin E turnover is normally approximately 1 h in sea urchin zygotes, the different behaviors of centrosomes during G1 and S phase arrests could be due to differential losses of cyclin E and its associated kinase activities at these two arrest points. To better understand the mechanisms that limit centrosome duplication, we characterize the levels of cyclin E and its associated kinase activity at the S phase and G1 arrest points. We first demonstrate that cyclin E/cdk2 kinase activity is required for centrosome duplication and reduplication in sea urchin zygotes. Next we find that cyclin E levels and cyclin E/cdk2 kinase activities are both constitutively and equivalently elevated during both the S phase and G1 arrests. This indicates that centrosome duplication during the G1 arrest is limited by a block to reduplication under conditions permissive for duplication. The cytoplasmic conditions of S phase, however, abrogate this block to reduplication.


Asunto(s)
Centrosoma/metabolismo , Ciclina E/metabolismo , Erizos de Mar/metabolismo , Cigoto/metabolismo , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Centrosoma/efectos de los fármacos , Centrosoma/enzimología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Fase G1/efectos de los fármacos , Purinas/farmacología , Roscovitina , Fase S/efectos de los fármacos , Erizos de Mar/citología , Erizos de Mar/efectos de los fármacos , Erizos de Mar/enzimología , Cigoto/citología , Cigoto/efectos de los fármacos , Cigoto/enzimología
15.
Mol Biol Cell ; 29(22): 2632-2643, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30133342

RESUMEN

When untransformed human cells spend >1.5 h in prometaphase under standard culture conditions, all daughters arrest in G1 despite normal division of their mothers. We investigate what happens during prolonged prometaphase that leads to daughter cell arrest in the absence of DNA damage. We find that progressive loss of anti-apoptotic MCL-1 activity and oxidative stress act in concert to partially activate the apoptosis pathway, resulting in the delayed death of some daughters and senescence for the rest. At physiological oxygen levels, longer prometaphase durations are needed for all daughters to arrest. Partial activation of apoptosis during prolonged prometaphase leads to persistent caspase activity, which activates the kinase cascade mediating the post-mitotic activation of p38. This in turn activates p53, and the consequent expression of p21stops the cell cycle. This mechanism can prevent cells suffering intractable mitotic defects, which modestly prolong mitosis but allow its completion without DNA damage, from producing future cell generations that are susceptible to the evolution of a transformed phenotype.


Asunto(s)
Apoptosis , Prometafase , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oxígeno/farmacología , Prometafase/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Dis Model Mech ; 10(12): 1381-1389, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29259023

RESUMEN

For hundreds of years, biologists have studied accessible organisms such as garden peas, sea urchins collected at low tide, newt eggs, and flies circling rotten fruit. These organisms help us to understand the world around us, attracting and inspiring each new generation of biologists with the promise of mystery and discovery. Time and time again, what we learn from such simple organisms has emphasized our common biological origins by proving to be applicable to more complex organisms, including humans. Yet, biologists are increasingly being tasked with developing applications from the known, rather than being allowed to follow a path to discovery of the as yet unknown. Here, we provide examples of important lessons learned from research using selected non-vertebrate organisms. We argue that, for the purpose of understanding human disease, simple organisms cannot and should not be replaced solely by human cell-based culture systems. Rather, these organisms serve as powerful discovery tools for new knowledge that could subsequently be tested for conservation in human cell-based culture systems. In this way, curiosity-driven biological research in simple organisms has and will continue to pay huge dividends in both the short and long run for improving the human condition.


Asunto(s)
Modelos Animales , Investigación , Animales , Pruebas Genéticas , Modelos Biológicos
17.
Cilia ; 5(1): 20, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27602205

RESUMEN

Centriole structure and function in the sea urchin zygote parallel those in mammalian somatic cells. Here, I briefly introduce the properties and attributes of the sea urchin system that make it an attractive platform for the study of centrosome and centriole duplication. These attributes apply to all echinoderms readily available from commercial suppliers: sea urchins, sand dollars, and starfish. I list some of the practical aspects of the system that make it a cost- and time-effective system for experimental work and then list properties that are a "tool kit" that can be used to conduct studies that would not be practical, or in some cases not possible, with mammalian somatic cells. Since centrioles organize and localize the pericentriolar material that nucleates the astral arrays of microtubules (Bobinnec et al. in J Cell Biol 143(6):1575-1589, 1998), the pattern of aster duplication over several cell cycles can be used as a reliable measure for centriole duplication (Sluder and Rieder in J Cell Biol 100(3):887-896, 1985). Descriptions of the methods my laboratory has used to handle and image echinoderm zygotes are reviewed in Sluder et al. (Methods Cell Biol 61:439-472, 1999). Also included is a bibliography of papers that describe additional methods.

18.
J Cell Biol ; 214(2): 143-53, 2016 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-27432896

RESUMEN

Precise regulation of centrosome number is critical for accurate chromosome segregation and the maintenance of genomic integrity. In nontransformed cells, centrosome loss triggers a p53-dependent surveillance pathway that protects against genome instability by blocking cell growth. However, the mechanism by which p53 is activated in response to centrosome loss remains unknown. Here, we have used genome-wide CRISPR/Cas9 knockout screens to identify a USP28-53BP1-p53-p21 signaling axis at the core of the centrosome surveillance pathway. We show that USP28 and 53BP1 act to stabilize p53 after centrosome loss and demonstrate this function to be independent of their previously characterized role in the DNA damage response. Surprisingly, the USP28-53BP1-p53-p21 signaling pathway is also required to arrest cell growth after a prolonged prometaphase. We therefore propose that centrosome loss or a prolonged mitosis activate a common signaling pathway that acts to prevent the growth of cells that have an increased propensity for mitotic errors.


Asunto(s)
Centrosoma/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Mitosis , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Secuencia de Bases , Sistemas CRISPR-Cas , Puntos de Control del Ciclo Celular , Línea Celular , Proliferación Celular , Daño del ADN , Técnicas de Inactivación de Genes , Humanos , Metafase , Estabilidad Proteica
19.
J Cell Biol ; 210(1): 63-77, 2015 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-26150389

RESUMEN

Centriole function has been difficult to study because of a lack of specific tools that allow persistent and reversible centriole depletion. Here we combined gene targeting with an auxin-inducible degradation system to achieve rapid, titratable, and reversible control of Polo-like kinase 4 (Plk4), a master regulator of centriole biogenesis. Depletion of Plk4 led to a failure of centriole duplication that produced an irreversible cell cycle arrest within a few divisions. This arrest was not a result of a prolonged mitosis, chromosome segregation errors, or cytokinesis failure. Depleting p53 allowed cells that fail centriole duplication to proliferate indefinitely. Washout of auxin and restoration of endogenous Plk4 levels in cells that lack centrioles led to the penetrant formation of de novo centrioles that gained the ability to organize microtubules and duplicate. In summary, we uncover a p53-dependent surveillance mechanism that protects against genome instability by preventing cell growth after centriole duplication failure.


Asunto(s)
Centriolos/genética , Inestabilidad Genómica , Proteína p53 Supresora de Tumor/fisiología , Puntos de Control del Ciclo Celular , Línea Celular , Proliferación Celular , Centriolos/fisiología , Segregación Cromosómica , Humanos , Microtúbulos/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/fisiología , Transporte de Proteínas
20.
BMC Cell Biol ; 5(1): 49, 2004 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-15617574

RESUMEN

BACKGROUND: A number of proteins accumulate in the spindle midzone and midbody of dividing animal cells. Besides proteins essential for cytokinesis, there are also components essential for interphase functions, suggesting that the spindle midzone and/or midbody may play a role in regulating the following cell cycle. RESULTS: We microsurgically severed NRK epithelial cells during anaphase or telophase, such that the spindle midzone/midbody was associated with only one of the daughter cells. Time-lapse recording of cells severed during early anaphase indicated that the cell with midzone underwent cytokinesis-like cortical contractions and progressed normally through the interphase, whereas the cell without midzone showed no cortical contraction and an arrest or substantial delay in the progression of interphase. Similar microsurgery during telophase showed a normal progression of interphase for both daughter cells with or without the midbody. Microsurgery of anaphase cells treated with cytochalasin D or nocodazole indicated that interphase progression was independent of cortical ingression but dependent on microtubules. CONCLUSIONS: We conclude that the mitotic spindle is involved in not only the separation of chromosomes but also the regulation of cell cycle. The process may involve activation of components in the spindle midzone that are required for the cell cycle, and/or degradation of components that are required for cytokinesis but may interfere with the cell cycle.


Asunto(s)
Anafase , Ciclo Celular , Huso Acromático/fisiología , Animales , Línea Celular , Células Epiteliales/citología , Células Epiteliales/ultraestructura , Interfase , Microcirugia , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA