Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38700016

RESUMEN

Myosin phosphatase targeting subunit1 (MYPT1) is a critical subunit of myosin phosphatase (MP), which brings PP1Cδ phosphatase and its substrate together. We previously showed that MYPT1 depletion resulted in oblique chromatid segregation. Therefore, we hypothesized that MYPT1 may control microtubule-dependent motor activity. Dynein, a minus-end microtubule motor, is known to be involved in mitotic spindle assembly. We thus examined whether MYPT1 and dynein may interact. Proximity ligation assay and co-immunoprecipitation revealed that MYPT1 and dynein intermediate chain (DIC) were associated. We found that DIC phosphorylation is increased in MYPT1-depleted cells in vivo, and that MP was able to dephosphorylate DIC in vitro. MYPT1 depletion also altered the localization and motility of Rab7-containing vesicles. MYPT1-depletion dispersed the perinuclear Rab7 localization to the peripheral in interphase cells. The dispersed Rab7 localization was rescued by microinjection of a constitutively active, truncated MYPT1 mutant, supporting that MP is responsible for the altered Rab7 localization. Analyses of Rab7 vesicle trafficking also revealed that minus-end transport was reduced in MYPT1-depleted cells. These results suggest an unexpected role of MP: MP controls dynein activity in both mitotic and interphase cells, possibly by dephosphorylating dynein subunits including DIC.

2.
J Med Internet Res ; 26: e54645, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38657229

RESUMEN

BACKGROUND: Understanding patient preference regarding taking tablet or capsule formulations plays a pivotal role in treatment efficacy and adherence. Therefore, these preferences should be taken into account when designing formulations and prescriptions. OBJECTIVE: This study investigates the factors affecting patient preference in patients who have difficulties swallowing large tablets or capsules and aims to identify appropriate sizes for tablets and capsules. METHODS: A robust data set was developed based on a questionnaire survey conducted from December 1, 2022, to December 7, 2022, using the harmo smartphone app operated by harmo Co, Ltd. The data set included patient input regarding their tablet and capsule preferences, personal health records (including dispensing history), and drug formulation information (available from package inserts). Based on the medication formulation information, 6 indices were set for each of the tablets or capsules that were considered difficult to swallow owing to their large size and concomitant tablets or capsules (used as controls). Receiver operating characteristic (ROC) analysis was used to evaluate the performance of each index. The index demonstrating the highest area under the curve of the ROC was selected as the best index to determine the tablet or capsule size that leads to swallowing difficulties. From the generated ROCs, the point with the highest discriminative performance that maximized the Youden index was identified, and the optimal threshold for each index was calculated. Multivariate logistic regression analysis was performed to identify the risk factors contributing to difficulty in swallowing oversized tablets or capsules. Additionally, decision tree analysis was performed to estimate the combined risk from several factors, using risk factors that were significant in the multivariate logistic regression analysis. RESULTS: This study analyzed 147 large tablets or capsules and 624 control tablets or capsules. The "long diameter + short diameter + thickness" index (with a 21.5 mm threshold) was identified as the best indicator for causing swallowing difficulties in patients. The multivariate logistic regression analysis (including 132 patients with swallowing difficulties and 1283 patients without) results identified the following contributory risk factors: aged <50 years (odds ratio [OR] 1.59, 95% CI 1.03-2.44), female (OR 2.54, 95% CI 1.70-3.78), dysphagia (OR 3.54, 95% CI 2.22-5.65), and taking large tablets or capsules (OR 9.74, 95% CI 5.19-18.29). The decision tree analysis results suggested an elevated risk of swallowing difficulties for patients with taking large tablets or capsules. CONCLUSIONS: This study identified the most appropriate index and threshold for indicating that a given tablet or capsule size will cause swallowing difficulties, as well as the contributory risk factors. Although some sampling biases (eg, only including smartphone users) may exist, our results can guide the design of patient-friendly formulations and prescriptions, promoting better medication adherence.


Asunto(s)
Cápsulas , Registros Electrónicos de Salud , Comprimidos , Humanos , Femenino , Masculino , Persona de Mediana Edad , Adulto , Anciano , Registros de Salud Personal , Trastornos de Deglución , Deglución , Encuestas y Cuestionarios , Prioridad del Paciente/estadística & datos numéricos
3.
J Cell Sci ; 133(24)2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33376154

RESUMEN

The centrosome, which consists of centrioles and pericentriolar material (PCM), becomes mature and assembles mitotic spindles by increasing the number of microtubules (MTs) emanating from the PCM. Among the molecules involved in centrosome maturation, Cep192 and Aurora A (AurA, also known as AURKA) are primarily responsible for recruitment of γ-tubulin and MT nucleators, whereas pericentrin (PCNT) is required for PCM organization. However, the role of Cep215 (also known as CDK5RAP2) in centrosome maturation remains elusive. Cep215 possesses binding domains for γ-tubulin, PCNT and MT motors that transport acentrosomal MTs towards the centrosome. We identify a mitosis-specific centrosome-targeting domain of Cep215 (215N) that interacts with Cep192 and phosphorylated AurA (pAurA). Cep192 is essential for targeting 215N to centrosomes, and centrosomal localization of 215N and pAurA is mutually dependent. Cep215 has a relatively minor role in γ-tubulin recruitment to the mitotic centrosome. However, it has been shown previously that this protein is important for connecting mitotic centrosomes to spindle poles. Based on the results of rescue experiments using versions of Cep215 with different domain deletions, we conclude that Cep215 plays a role in maintaining the structural integrity of the spindle pole by providing a platform for the molecules involved in centrosome maturation.


Asunto(s)
Aurora Quinasa A , Mitosis , Aurora Quinasa A/genética , Proteínas de Ciclo Celular/genética , Centrosoma , Proteínas Cromosómicas no Histona/genética , Células HeLa , Humanos , Proteínas del Tejido Nervioso , Huso Acromático/genética , Tubulina (Proteína)/genética
4.
FASEB J ; 33(7): 8363-8374, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30964711

RESUMEN

Cellular checkpoints controlling entry into mitosis monitor the integrity of the DNA and delay mitosis onset until the alteration is fully repaired. However, this canonical response can weaken, leading to a spontaneous bypass of the checkpoint, a process referred to as checkpoint adaptation. Here, we have investigated the contribution of microcephalin 1 (MCPH1), mutated in primary microcephaly, to the decatenation checkpoint, a less-understood G2 pathway that delays entry into mitosis until chromosomes are properly disentangled. Our results demonstrate that, although MCPH1 function is dispensable for activation and maintenance of the decatenation checkpoint, it is required for the adaptive response that bypasses the topoisomerase II inhibition----mediated G2 arrest. MCPH1, however, does not confer adaptation to the G2 arrest triggered by the ataxia telangiectasia mutated- and ataxia telangiectasia and rad3 related-based DNA damage checkpoint. In addition to revealing a new role for MCPH1 in cell cycle control, our study provides new insights into the genetic requirements that allow cellular adaptation to G2 checkpoints, a process that remains poorly understood.-Arroyo, M., Kuriyama, R., Guerrero, I., Keifenheim, D., Cañuelo, A., Calahorra, J., Sánchez, A., Clarke, D. J., Marchal, J. A. MCPH1 is essential for cellular adaptation to the G2-phase decatenation checkpoint.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Transformada , Proteínas del Citoesqueleto/genética , Humanos
5.
Mol Cell Biol ; 36(21): 2668-2680, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27528616

RESUMEN

Chibby1 (Cby1) is a small, conserved coiled-coil protein that localizes to centrioles/basal bodies and plays a crucial role in the formation and function of cilia. During early stages of ciliogenesis, Cby1 is required for the efficient recruitment of small vesicles at the distal end of centrioles to facilitate basal body docking to the plasma membrane. Here, we identified family with sequence similarity 92, member A (FAM92A) and FAM92B, which harbor predicted lipid-binding BAR domains, as novel Cby1-interacting partners using tandem affinity purification and mass spectrometry. We found that in cultured cell lines, FAM92A colocalizes with Cby1 at the centrioles/basal bodies of primary cilia, while FAM92B is undetectable. In airway multiciliated cells, both FAM92A and -92B colocalize with Cby1 at the base of cilia. Notably, the centriolar localization of FAM92A and -92B depends largely on Cby1. Knockdown of FAM92A in RPE1 cells impairs ciliogenesis. Consistent with the membrane-remodeling properties of BAR domains, FAM92A and -92B in cooperation with Cby1 induce deformed membrane-like structures containing the small GTPase Rab8 in cultured cells. Our results therefore suggest that FAM92 proteins interact with Cby1 to promote ciliogenesis via regulation of membrane-remodeling processes.


Asunto(s)
Proteínas Portadoras/metabolismo , Cilios/metabolismo , Morfogénesis , Proteínas Nucleares/metabolismo , Proteínas/química , Proteínas/metabolismo , Animales , Cuerpos Basales/metabolismo , Centriolos/metabolismo , Células Epiteliales/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Dominios Proteicos , Multimerización de Proteína
6.
J Clin Endocrinol Metab ; 100(6): E883-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25901990

RESUMEN

CONTEXT: Most epigenetic studies in diabetes compare normal cells in "high glucose" (HG) to cells in "normal glucose" (NG) and cells returned from HG to NG. Here we challenge this approach. OBJECTIVE: The objective was to determine whether there were differences in gene expression in skin fibroblasts of monozygotic twins (MZT) discordant for type 1 diabetes (T1D). DESIGN: Skin fibroblasts were grown in NG (5.5 mmol/L) and HG (25 mmol/L) for multiple passages. SETTING: This study was conducted at the University of Minnesota. PATIENTS: Patients were nine MZT pairs discordant for T1D. MAIN OUTCOME MEASURE(S): Gene expression was assessed by mRNA-Seq, using the Illumina HiSeq 2000 instrument. Pathway analysis tested directionally consistent group differences within the Kyoto Encyclopedia of Genes and Genomes pathways. RESULTS: A total of 3308 genes were differentially expressed between NG and HG in T1D MZT vs 889 in non-T1D twins. DNA replication, proteasome, cell cycle, base excision repair, homologous recombination, pyrimidine metabolism, and spliceosome pathways had overrepresented genes with increased expression in T1D twins with P values ranging from 7.21 × 10(-10) to 1.39 × 10(-4). In a companion article, we demonstrate that these pathway changes are related to diabetic nephropathy risk. There were no pathways statistically significant differently expressed in nondiabetic twins in HG vs NG. CONCLUSIONS: In vivo exposure to diabetes alters cells in a manner that markedly changes their in vitro responses to HG. These results highlight the importance of using cells directly derived from diabetic patients for studies examining the effects of HG in diabetes.


Asunto(s)
Glucemia/genética , Diabetes Mellitus Tipo 1/genética , Hiperglucemia/genética , Adulto , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Persona de Mediana Edad , Cultivo Primario de Células , Piel/efectos de los fármacos , Piel/metabolismo , Gemelos Monocigóticos
7.
J Cell Biol ; 207(1): 123-37, 2014 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-25313408

RESUMEN

Airway multiciliated epithelial cells play crucial roles in the mucosal defense system, but their differentiation process remains poorly understood. Mice lacking the basal body component Chibby (Cby) exhibit impaired mucociliary transport caused by defective ciliogenesis, resulting in chronic airway infection. In this paper, using primary cultures of mouse tracheal epithelial cells, we show that Cby facilitates basal body docking to the apical cell membrane through proper formation of ciliary vesicles at the distal appendage during the early stages of ciliogenesis. Cby is recruited to the distal appendages of centrioles via physical interaction with the distal appendage protein CEP164. Cby then associates with the membrane trafficking machinery component Rabin8, a guanine nucleotide exchange factor for the small guanosine triphosphatase Rab8, to promote recruitment of Rab8 and efficient assembly of ciliary vesicles. Thus, our study identifies Cby as a key regulator of ciliary vesicle formation and basal body docking during the differentiation of airway ciliated cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Cilios/metabolismo , Células Epiteliales/citología , Proteínas de Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Mucosa Respiratoria/citología , Secuencias de Aminoácidos/genética , Animales , Cuerpos Basales/fisiología , Proteínas Portadoras/genética , Diferenciación Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Centriolos/fisiología , Cilios/genética , Quinasas del Centro Germinal , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microtúbulos/genética , Depuración Mucociliar/genética , Naftalenos , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Unión al GTP rab/metabolismo
8.
Mol Biol Cell ; 24(17): 2645-54, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864714

RESUMEN

Centrosomes are key microtubule-organizing centers that contain a pair of centrioles, conserved cylindrical, microtubule-based structures. Centrosome duplication occurs once per cell cycle and relies on templated centriole assembly. In many animal cells this process starts with the formation of a radially symmetrical cartwheel structure. The centrosomal protein Cep135 localizes to this cartwheel, but its role in vertebrates is not well understood. Here we examine the involvement of Cep135 in centriole function by disrupting the Cep135 gene in the DT40 chicken B-cell line. DT40 cells that lack Cep135 are viable and show no major defects in centrosome composition or function, although we note a small decrease in centriole numbers and a concomitant increase in the frequency of monopolar spindles. Furthermore, electron microscopy reveals an atypical structure in the lumen of Cep135-deficient centrioles. Centrosome amplification after hydroxyurea treatment increases significantly in Cep135-deficient cells, suggesting an inhibitory role for the protein in centrosome reduplication during S-phase delay. We propose that Cep135 is required for the structural integrity of centrioles in proliferating vertebrate cells, a role that also limits centrosome amplification in S-phase-arrested cells.


Asunto(s)
Proteínas Aviares/metabolismo , Proteínas Portadoras/fisiología , Centriolos/metabolismo , Centrosoma/metabolismo , Centrosoma/ultraestructura , Animales , Proteínas Aviares/genética , Proteínas Portadoras/genética , Ciclo Celular/genética , División Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Centriolos/genética , Centrosoma/química , Pollos , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Marcación de Gen , Microtúbulos/genética , Microtúbulos/metabolismo , Mitosis , Fase S/genética , Fase S/fisiología
9.
PLoS One ; 7(7): e41077, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22911743

RESUMEN

The mother centriole of the centrosome is distinguished from immature daughter centrioles by the presence of accessory structures (distal and subdistal appendages), which play an important role in the organization of the primary cilium in quiescent cells. Primary cilia serve as sensory organelles, thus have been implicated in mediating intracellular signal transduction pathways. Here we report that Chibby (Cby), a highly conserved antagonist of the Wnt/ß-catenin pathway, is a centriolar component specifically located at the distal end of the mother centriole and essential for assembly of the primary cilium. Cby appeared as a discrete dot in the middle of a ring-like structure revealed by staining with a distal appendage component of Cep164. Cby interacted with one of the appendage components, Cenexin (Cnx), which thereby abrogated the inhibitory effect of Cby on ß-catenin-mediated transcriptional activation in a dose-dependent manner. Cby and Cnx did not precisely align, as Cby was detected at a more distal position than Cnx. Cnx emerged earlier than Cby during the cell cycle and was required for recruitment of Cby to the mother centriole. However, Cby was dispensable for Cnx localization to the centriole. During massive centriogenesis in in vitro cultured mouse tracheal epithelial cells, Cby and Cnx were expressed in a similar pattern, which was coincident with the expression of Foxj1. Our results suggest that Cby plays an important role in organization of both primary and motile cilia in collaboration with Cnx.


Asunto(s)
Proteínas Portadoras/metabolismo , Centriolos/metabolismo , Cilios/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Ciclo Celular , Diferenciación Celular , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Ratones , Unión Proteica , Transporte de Proteínas , Activación Transcripcional
10.
Cytoskeleton (Hoboken) ; 68(8): 446-58, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21766470

RESUMEN

To study the mechanism of centrosome duplication in cycling cells, we established a novel system of multiple centrosome formation in two types of cells: CHO cells treated with RO3306, a Cyclin-dependent kinase 1 (Cdk1) inhibitor and DT40 cells, in which Cdks were knocked out by chemical genetics. Cdk1-inactivated cells initiated DNA replication and centrosome duplication at the onset of S phase. They became arrested at the end of G2, but the centrosome cycle continued to produce supernumerary centrioles/centrosomes without DNA endoreplication in those cells. Centrosomes were amplified in a highly synchronous and reproducible manner: all of them were located next to the nucleus and spread widely apart from each other with several µm in distance. Double knockout of Cdk1 and Cdk2 caused cell cycle arrest at G1/S and centrosomes were no longer duplicated. However, cells continued to grow and increased their volume over 10-fold during 48 hr of culture. Centrosome components, including γ-tubulin and Cep135, were synthesized and accumulated during the arrest, allowing rapid centrosome multiplication upon recovery from the cell cycle arrest or expression of exogenous Plk4 in G1/S cells. Thus centrosome amplification results from the discoordination of the centrosome cycle from the progression of other cell cycle events, which is controlled by different levels of Cdk activities.


Asunto(s)
Centrosoma/fisiología , Quinasas Ciclina-Dependientes/metabolismo , Animales , Células CHO , Ciclo Celular/fisiología , Línea Celular , Centrosoma/enzimología , Cricetinae , Cricetulus , Humanos , Microscopía de Contraste de Fase
11.
EMBO J ; 30(11): 2233-45, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21522128

RESUMEN

Cyclin-dependent kinase 1 (Cdk1) is thought to trigger centrosome separation in late G2 phase by phosphorylating the motor protein Eg5 at Thr927. However, the precise control mechanism of centrosome separation remains to be understood. Here, we report that in G2 phase polo-like kinase 1 (Plk1) can trigger centrosome separation independently of Cdk1. We find that Plk1 is required for both C-Nap1 displacement and for Eg5 localization on the centrosome. Moreover, Cdk2 compensates for Cdk1, and phosphorylates Eg5 at Thr927. Nevertheless, Plk1-driven centrosome separation is slow and staggering, while Cdk1 triggers fast movement of the centrosomes. We find that actin-dependent Eg5-opposing forces slow down separation in G2 phase. Strikingly, actin depolymerization, as well as destabilization of interphase microtubules (MTs), is sufficient to remove this obstruction and to speed up Plk1-dependent separation. Conversely, MT stabilization in mitosis slows down Cdk1-dependent centrosome movement. Our findings implicate the modulation of MT stability in G2 and M phase as a regulatory element in the control of centrosome separation.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular , Centrosoma/metabolismo , Cinesinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Línea Celular , Humanos , Quinasa Tipo Polo 1
12.
Cell Cycle ; 8(17): 2802-9, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19652536

RESUMEN

Cdk2 was once believed to play an essential role in cell cycle progression, but cdk2(-/-) mice have minimal phenotypic abnormalities. In this study, we examined the role of cdk2 in hepatocyte proliferation, centrosome duplication and survival. Cdk2(-/-) hepatocytes underwent mitosis and had normal centrosome content after mitogen stimulation. Unlike wild-type cells, cdk2(-/-) liver cells failed to undergo centrosome overduplication in response to ectopic cyclin D1 expression. After mitogen stimulation in culture or partial hepatectomy in vivo, cdk2(-/-) hepatocytes demonstrated diminished proliferation. Cyclin D1 is a key mediator of cell cycle progression in hepatocytes, and transient expression of this protein is sufficient to promote robust proliferation of these cells in vivo. In cdk2(-/-) mice and animals treated with the cdk2 inhibitor seliciclib, cyclin D1 failed to induce hepatocyte cell cycle progression. Surprisingly, cdk2 ablation or inhibition led to massive hepatocyte and animal death following cyclin D1 transfection. In a transgenic model of chronic hepatic cyclin D1 expression, seliciclib induced hepatocyte injury and animal death, suggesting that cdk2 is required for survival of cyclin D1-expressing cells even in the absence of substantial proliferation. In conclusion, our studies demonstrate that cdk2 plays a role in liver regeneration. Furthermore, it is essential for centrosome overduplication, proliferation and survival of hepatocytes that aberrantly express cyclin D1 in vivo. These studies suggest that cdk2 may warrant further investigation as a target for therapy of liver tumors with constitutive cyclin D1 expression.


Asunto(s)
Ciclo Celular , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/fisiología , Hepatocitos/enzimología , Animales , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Centrosoma/metabolismo , Ciclina A/metabolismo , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Regeneración Hepática , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Purinas/farmacología , Roscovitina , Factores de Tiempo , Transfección
13.
J Cell Sci ; 122(Pt 12): 2014-23, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19454482

RESUMEN

Cancer cells frequently induce aberrant centrosomes, which have been implicated in cancer initiation and progression. Human colorectal cancer cells, HCT116, contain aberrant centrioles composed of disorganized cylindrical microtubules and displaced appendages. These cells also express unique centrosome-related structures associated with a subset of centrosomal components, including gamma-tubulin, centrin and PCM1. During hydroxyurea treatment, these abnormal structures become more abundant and undergo a change in shape from small dots to elongated fibers. Although gamma-tubulin seems to exist as a ring complex, the abnormal structures do not support microtubule nucleation. Several lines of evidence suggest that the fibers correspond to a disorganized form of centriolar microtubules. Plk4, a mammalian homolog of ZYG-1 essential for initiation of centriole biogenesis, is not associated with the gamma-tubulin-specific abnormal centrosomes. The amount of Plk4 at each centrosome was less in cells with abnormal centrosomes than cells without gamma-tubulin-specific abnormal centrosomes. In addition, the formation of abnormal structures was abolished by expression of exogenous Plk4, but not SAS6 and Cep135/Bld10p, which are downstream regulators required for the organization of nine-triplet microtubules. These results suggest that HCT116 cells fail to organize the ninefold symmetry of centrioles due to insufficient Plk4.


Asunto(s)
Centriolos/patología , Neoplasias Colorrectales/patología , Células HCT116 , Proteínas Serina-Treonina Quinasas/metabolismo , Tubulina (Proteína)/metabolismo , Centriolos/efectos de los fármacos , Centriolos/metabolismo , Centriolos/ultraestructura , Neoplasias Colorrectales/metabolismo , Humanos , Hidroxiurea/farmacología , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Nocodazol/farmacología , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Moduladores de Tubulina/farmacología , Regulación hacia Arriba
14.
Cell Motil Cytoskeleton ; 66(8): 588-96, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19402176

RESUMEN

Centriole assembly is initiated by Plk4, a Polo-like kinase 4, which causes the recruitment of downstream regulators, such as SAS6 and SAS4, to a nascent centriole. Simultaneous expression of Plk4, SAS6 and SAS4 in CHO cells resulted in the formation of massive fibrogranular aggregates of various sizes and shapes. These aggregates were surrounded by dense particles of about 70 nm in diameter, similar to the centriolar satellite that has been observed around the centrosome in normal cycling cells. Within the fibrillar material, ring-like structures appeared and eventually differentiated into centrioles by association with short microtubule bundles. Centrioles were also assembled around a parent centriole in a cluster, a configuration that has been described as a "flower structure" formation [Kleylein-Sohn et al.,2007]. This pattern of centriole duplication is reminiscent of the arrangement of new centrioles induced in normal ciliated trachea/oviduct cells by the centriole-dependent pathway, which was reported several decades ago [Sorokin,1968; Anderson and Brenner,1971; Dirksen,1971]. Prior to the production of hundreds of centrioles, these differentiating epithelial cells were also shown to induce a dense filamentous material similar to that detected in transfected CHO cells. These results suggest a common mechanism of centriole assembly regulated by Plk4 in both transfected cycling cells and normal ciliated epithelial cells undergoing differentiation. Cell Motil. Cytoskeleton 2009. (c) 2009 Wiley-Liss, Inc.


Asunto(s)
Centriolos/metabolismo , Cilios/metabolismo , Células Epiteliales/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Células CHO , Centriolos/genética , Centriolos/ultraestructura , Cilios/fisiología , Cilios/ultraestructura , Cricetinae , Cricetulus , Células Epiteliales/ultraestructura , Femenino , Humanos , Ratones , Microscopía Electrónica de Transmisión , Oviductos/citología , Proteínas Serina-Treonina Quinasas/genética
15.
J Cell Sci ; 120(Pt 14): 2444-53, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17606999

RESUMEN

Centrosome duplication is tightly coupled with the cell cycle and neither too many nor too few centrosomes are induced in a normal cell. To study how centrosome assembly is regulated, we analyzed the abnormal process of multiple centrosome replications in Chinese hamster ovary (CHO) cells induced by hydroxyurea (HU), which is known to uncouple the centrosome cycle from the cell cycle. Green fluorescent protein (GFP)-tagged centrin2 expressed in CHO cells labels both centrioles and the pericentriolar material (PCM). Counting fluorescent spots of GFP-centrin in synchronized cells showed that in G(1)/S-arrested cells, centrioles are initially duplicated in a template manner. Further treatment with HU overrides the suppression of excess centriole/centrosome replication in a cell where the full complement of centrioles/centrosomes already exists. Time-lapse fluorescence microscopy revealed that small centrin-containing foci emerged in the cytoplasm during HU treatment. These foci are surrounded by a PCM cloud and their number continuously increases as cells are exposed to HU for longer periods of time. Both the centrosome and cytoplasmic foci are highly mobile, continuously changing their position in a manner dependent on microtubules/microtubule dynamics. The centrosome number increases as small foci grow in size and resolve into recognizable centrosomes. As this occurs in a random fashion, the cells arrested longer with HU induced highly heterogeneous numbers of centrosomes.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Centrosoma/metabolismo , Animales , Células CHO , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Cricetinae , Cricetulus , Femenino , Fluoroinmunoensayo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hidroxiurea/farmacología , Mitosis , Proteínas Recombinantes de Fusión/metabolismo , Huso Acromático/metabolismo
16.
Eur J Cell Biol ; 85(12): 1233-40, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17000024

RESUMEN

We addressed the role of the G-protein regulatory (GPR) motif-containing Leu-Gly-Asn-enriched protein (LGN) and G-proteins (Gialpha3) in the positioning of the spindle pole during mammalian cell division. Immunocytochemistry indicated that both LGN and Gialpha3 co-localized at the spindle pole and at the midbody and the cell cortex during the different phases of mitosis. In marked contrast to the positioning of the spindle pole at metaphase midway between the cell cortex and the metaphase plate, the spindle pole was juxtaposed with the cell cortex at metaphase following increased expression of Gialpha3 and LGN. This repositioning of the spindle pole required the interaction of LGN with Gialpha. The influence of LGN and Gialpha3 on the cortical positioning of the spindle pole likely reflects either stronger pulling forces on the spindle pole exerted from the cell cortex or increased pushing forces exerted on the spindle pole from the mitotic spindle indicating that these events are regulated by GPR motif-containing proteins and G-proteins independent of asymmetry.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Proteínas de Unión al GTP/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Metafase/fisiología , Mitosis/fisiología , Huso Acromático/fisiología , Secuencias de Aminoácidos/fisiología , Animales , Asparagina/análisis , Células CHO , Cricetinae , Cricetulus , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/análisis , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Glicina/análisis , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Leucina/análisis , Metafase/genética , Mitosis/genética
17.
Mol Cell Biol ; 26(22): 8316-35, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16966375

RESUMEN

Outer dense fiber 2 (Odf2) was initially identified as a major component of sperm tail cytoskeleton and later was suggested to be a widespread component of centrosomal scaffold that preferentially associates with the appendages of the mother centrioles in somatic cells. Here we report the identification of two Odf2-related centrosomal components, hCenexin1 and hCenexin1 variant 1, that possess a unique C-terminal extension. Our results showed that hCenexin1 is the major isoform expressed in HeLa cells, whereas hOdf2 is not detectably expressed. Mammalian polo-like kinase 1 (Plk1) is critical for proper mitotic progression, and its association with the centrosome is important for microtubule nucleation and function. Interestingly, depletion of hCenexin1 by RNA interference (RNAi) delocalized Plk1 from the centrosomes and the C-terminal extension of hCenexin1 was crucial to recruit Plk1 to the centrosomes through a direct interaction with the polo-box domain of Plk1. Consistent with these findings, the hCenexin1 RNAi cells exhibited weakened gamma-tubulin localization and chromosome segregation defects. We propose that hCenexin1 is a critical centrosomal component whose C-terminal extension is required for proper recruitment of Plk1 and other components crucial for normal mitosis. Our results further suggest that the anti-Odf2 immunoreactive centrosomal antigen previously detected in non-germ line cells is likely hCenexin1.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Centrosoma/metabolismo , Proteínas de Choque Térmico/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Células CHO , Ciclo Celular , Proteínas de Ciclo Celular/fisiología , Línea Celular Tumoral , Segregación Cromosómica , Cricetinae , Células HeLa , Proteínas de Choque Térmico/genética , Humanos , Datos de Secuencia Molecular , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , ARN Interferente Pequeño , Homología de Secuencia de Aminoácido , Huso Acromático , Transfección , Quinasa Tipo Polo 1
18.
Cell Motil Cytoskeleton ; 62(4): 195-209, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16240430

RESUMEN

Lecudina tuzetae is a parasitic protozoan (Gregarine, Apicomplexa) living in the intestine of a marine polychaete annelid, Nereis diversicolor. Using electron and fluorescence microscopy, we have characterized the dynamic changes in microtubule organization during the sexual phase of the life cycle. The gametocyst excreted from the host worm into seawater consists of two (one male and one female) gamonts in which cortical microtubule arrays are discernible. Each gamont undergoes multiple nuclear divisions without cytokinesis, resulting in the formation of large multinucleate haploid cells. After cellularization, approximately 1000 individual gametes are produced from each gamont within 24 h. Female gametes are spherical and contain interphase cytoplasmic microtubule arrays emanating from a gamma-tubulin-containing site. In male gametes, both interphase microtubules and a flagellum with "6 + 0" axonemal microtubules extend from the same microtubule-organizing site. At the beginning of spore formation, each zygote secretes a wall to form a sporocyst. Following meiotic and mitotic divisions, each sporocyst gives rise to eight haploid cells that ultimately differentiate into sporozoites. The ovoid shaped sporocyst is asymmetric and forms at least two distinctive microtubule arrays: spindle microtubules and microtubule bundles originating from the protruding apical end corresponding to the dehiscence pole of the sporocyst. Because antibodies raised against mammalian centrosome components, such as gamma-tubulin, pericentrin, Cep135, and mitosis-specific phosphoproteins, react strongly with the microtubule-nucleating sites of Lecudina, this protozoan is likely to share common centrosomal antigens with higher eukaryotes.


Asunto(s)
Apicomplexa/crecimiento & desarrollo , Apicomplexa/fisiología , Centro Organizador de los Microtúbulos/ultraestructura , Microtúbulos/ultraestructura , Poliquetos/parasitología , Tubulina (Proteína)/metabolismo , Animales , Anticuerpos/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , División del Núcleo Celular , Centrosoma/metabolismo , Centrosoma/ultraestructura , Reacciones Cruzadas , Diploidia , Femenino , Fertilización , Flagelos/metabolismo , Flagelos/ultraestructura , Técnica del Anticuerpo Fluorescente , Células Germinativas/metabolismo , Células Germinativas/ultraestructura , Haploidia , Interacciones Huésped-Parásitos , Interfase , Estadios del Ciclo de Vida , Masculino , Meiosis , Microscopía Fluorescente , Centro Organizador de los Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitosis , Modelos Biológicos , Huso Acromático/metabolismo , Huso Acromático/ultraestructura , Cigoto/metabolismo , Cigoto/ultraestructura
19.
J Biol Chem ; 280(1): 768-76, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15509582

RESUMEN

In normal cells, cyclin D1 is induced by growth factors and promotes progression through the G(1) phase of the cell cycle. Cyclin D1 is also an oncogene that is thought to act primarily by bypassing the requirement for mitogens during the G(1) phase. Studies of clinical tumors have found that cyclin D1 overexpression is associated with chromosome abnormalities, although a causal effect has not been established in experimental systems. In this study, we found that transient expression of cyclin D1 in normal hepatocytes in vivo triggered dysplastic mitoses, accumulation of supernumerary centrosomes, abnormalities of the mitotic spindle, and marked chromosome changes within several days. This was associated with up-regulation of checkpoint genes p53 and p21 as well as hepatocyte apoptosis in the liver. Transient transfection of cyclin D1 also induced centrosome and mitotic spindle abnormalities in breast epithelial cells, suggesting that this may be a generalized effect. These results indicate that cyclin D1 can induce deregulation of the mitotic apparatus and aneuploidy, effects that could contribute to the role of this oncogene in malignancy.


Asunto(s)
Aneuploidia , Centrosoma/metabolismo , Ciclina D1/genética , Huso Acromático/metabolismo , Animales , Transformación Celular Neoplásica , Células Cultivadas , Ciclina D1/biosíntesis , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Masculino , Ratones , Ratones Endogámicos BALB C , Huso Acromático/ultraestructura
20.
J Cell Sci ; 117(Pt 15): 3233-46, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15199097

RESUMEN

Polo-like kinases and kinesin-like motor proteins are among the many proteins implicated in the execution of cytokinesis. Polo-like-kinase 1 (Plk1) interacts with the mitotic kinesin-like motor protein CHO1/MKLP-1 during anaphase and telophase, and CHO1/MKLP-1 is a Plk1 substrate in vitro. Here, we explore the molecular interactions of these two key contributors to mitosis and cytokinesis. Using the transient transfection approach, we show that the C-terminus of Plk1 binds CHO1/MKLP-1 in a Polo-box-dependent manner and that the stalk domain of CHO1/MKLP-1 is responsible for its binding to Plk1. The stalk domain was found to localize with Plk1 to the mid-body, and Plk1 appears to be mislocalized in CHO1/MKLP-1-depleted cells during late mitosis. We showed that Ser904 and Ser905 are two major Plk1 phosphorylation sites. Using the vector-based RNA interference approach, we showed that depletion of CHO1/MKLP-1 causes the formation of multinucleate cells with more centrosomes, probably because of a defect in the early phase of cytokinesis. Overexpression of a non-Plk1-phosphorylatable CHO1 mutant caused cytokinesis defects, presumably because of dominant negative effect of the construct. Finally, CHO1-depletion-induced multinucleation could be partially rescued by co-transfection of a non-degradable hamster wild-type CHO1 construct, but not an unphosphorylatable mutant. These data provide more detailed information about the interaction between Plk1 and CHO1/MKLP-1, and the significance of this is discussed.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Quinasas/metabolismo , Animales , Sitios de Unión , Western Blotting , Células CHO , Células COS , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Cricetinae , Citocinesis , ADN/metabolismo , Fase G1 , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Fluorescente , Mitosis , Modelos Genéticos , Mutación , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas , ARN/química , Interferencia de ARN , Serina/química , Telofase , Factores de Tiempo , Transfección , Quinasa Tipo Polo 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...