Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell ; 81(22): 4722-4735.e5, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34626566

RESUMEN

Rapid protein degradation enables cells to quickly modulate protein abundance. Dysregulation of short-lived proteins plays essential roles in disease pathogenesis. A focused map of short-lived proteins remains understudied. Cycloheximide, a translational inhibitor, is widely used in targeted studies to measure degradation kinetics for short-lived proteins. Here, we combined cycloheximide chase assays with advanced quantitative proteomics to map short-lived proteins under translational inhibition in four human cell lines. Among 11,747 quantified proteins, we identified 1,017 short-lived proteins (half-lives ≤ 8 h). These short-lived proteins are less abundant, evolutionarily younger, and less thermally stable than other proteins. We quantified 103 proteins with different stabilities among cell lines. We showed that U2OS and HCT116 cells express truncated forms of ATRX and GMDS, respectively, which have lower stability than their full-length counterparts. This study provides a large-scale resource of human short-lived proteins under translational arrest, leading to untapped avenues of protein regulation for therapeutic interventions.


Asunto(s)
Proteínas/química , Proteoma , Proteómica/métodos , Alanina/análogos & derivados , Alanina/química , Línea Celular , Línea Celular Tumoral , Cicloheximida/química , Cicloheximida/farmacología , Fucosa/química , Geminina/química , Células HCT116 , Células HEK293 , Humanos , Péptidos/química , Análisis de Componente Principal , Biosíntesis de Proteínas , Proteínas/efectos de los fármacos , Control de Calidad , ARN Interferente Pequeño/metabolismo , Telómero/química
2.
Methods Mol Biol ; 2329: 291-309, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34085231

RESUMEN

The cell and cilia cycles are inextricably linked through the dual functions of the centrioles at both the basal body of cilia and at mitotic centrosomes. How cilia assembly and disassembly, either through slow resorption or rapid deciliation, are coordinated with cell cycle progression remains unclear in many cell types and developmental paradigms. Moreover, little is known about how additional cilia parameters including changes in ciliary length or frequency of distal tip shedding change with cell cycle stage. In order to explore these questions, we have developed the Arl13bCerulean-Fucci2a tricistronic cilia and cell cycle biosensor (Ford et al., Dev Cell 47:509-523.e7, 2018). This reporter allowed us to document the heterogeneity in ciliary behaviors during the cell cycle at a population level. Without the need for external stimuli, it revealed that in several cell types and in the developing embryo cilia persist beyond the G1/S checkpoint. Here, we describe the generation of stable cell lines expressing Arl13bCerulean-Fucci2a and open-source software to aid morphometric profiling of the primary cilium with cell cycle phases, including changes in cilium length. This resource will allow the investigation of multiple morphometric questions relating to cilia and cell cycle biology.


Asunto(s)
Técnicas Biosensibles/métodos , Cilios/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Células 3T3 , Animales , Ciclo Celular , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Geminina/química , Geminina/metabolismo , Humanos , Proteínas Luminiscentes/metabolismo , Ratones , Microscopía Confocal , Dominios Proteicos , Proteínas Recombinantes de Fusión/química , Proteína Fluorescente Roja
3.
PLoS Comput Biol ; 16(6): e1007812, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32497127

RESUMEN

Apoptotic cell death can be initiated through the extrinsic and intrinsic signaling pathways. While cell cycle progression promotes the responsiveness to intrinsic apoptosis induced by genotoxic stress or spindle poisons, this has not yet been studied conclusively for extrinsic apoptosis. Here, we combined fluorescence-based time-lapse monitoring of cell cycle progression and cell death execution by long-term time-lapse microscopy with sampling-based mathematical modeling to study cell cycle dependency of TRAIL-induced extrinsic apoptosis in NCI-H460/geminin cells. In particular, we investigated the interaction of cell death timing and progression of cell cycle states. We not only found that TRAIL prolongs cycle progression, but in reverse also that cell cycle progression affects the kinetics of TRAIL-induced apoptosis: Cells exposed to TRAIL in G1 died significantly faster than cells stimulated in S/G2/M. The connection between cell cycle state and apoptosis progression was captured by developing a mathematical model, for which parameter estimation revealed that apoptosis progression decelerates in the second half of the cell cycle. Similar results were also obtained when studying HCT-116 cells. Our results therefore reject the null hypothesis of independence between cell cycle progression and extrinsic apoptosis and, supported by simulations and experiments of synchronized cell populations, suggest that unwanted escape from TRAIL-induced apoptosis can be reduced by enriching the fraction of cells in G1 phase. Besides novel insight into the interrelation of cell cycle progression and extrinsic apoptosis signaling kinetics, our findings are therefore also relevant for optimizing future TRAIL-based treatment strategies.


Asunto(s)
Apoptosis , Ciclo Celular , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Transducción de Señal , Algoritmos , Teorema de Bayes , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Geminina/química , Células HCT116 , Humanos , Cinética , Modelos Estadísticos
4.
Cell Cycle ; 16(9): 830-840, 2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28379781

RESUMEN

DNA replication is rigorously controlled in cells to ensure that the genome duplicates exactly once per cell cycle. Geminin is a small nucleoprotein, which prevents DNA rereplication by directly binding to and inhibiting the DNA replication licensing factor, Cdt1. In this study, we have identified 2 Geminin genes, BmGeminin1 and BmGeminn2, in silkworm, Bombyx mori. These genes contain the Geminin conserved coiled-coil domain and are periodically localized in the nucleus during the S-G2 phase but are degraded at anaphase in mitosis. Both BmGeminin1 and BmGeminin2 are able to homodimerize and interact with BmCdt1 in cells. In addition, BmGeminin1 and BmGeminin2 can interact with each other. Overexpression of BmGeminin1 affects cell cycle progression: cell cycle is arrested in S phase, and RNA interference of BmGeminin1 leads to rereplication. In contrast, overexpression or knockdown of BmGeminin2 with RNAi did not significantly affect cell cycle, while more rereplication occurred when BmGeminin1 and BmGeminin2 together were knocked down in cells than when only BmGeminin1 was knocked down. These data suggest that both BmGeminin1 and BmGeminin2 are involved in the regulation of DNA replication. These findings provide insight into the function of Geminin and contribute to our understanding of the regulation mechanism of cell cycle in silkworm.


Asunto(s)
Bombyx/metabolismo , Replicación del ADN , Geminina/química , Geminina/metabolismo , Proteínas de Insectos/química , Proteínas de Insectos/metabolismo , Homología de Secuencia de Aminoácido , Secuencia de Aminoácidos , Animales , Ciclo Celular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Filogenia , Unión Proteica
5.
Biochimie ; 131: 115-127, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27702582

RESUMEN

DNA replicates in a timely manner with each cell division. Multiple proteins and factors are involved in the initiation of DNA replication including a dynamic interaction between Cdc10-dependent transcript (Cdt1) and Geminin (GMNN). A conformational change between GMNN-Cdt1 heterotrimer and heterohexamer complex is responsible for licensing or inhibition of the DNA replication. This molecular switch ensures a faithful DNA replication during each S phase of cell cycle. GMNN inhibits Cdt1-mediated minichromosome maintenance helicases (MCM) loading onto the chromatin-bound origin recognition complex (ORC) which results in the inhibition of pre-replication complex assembly. GMNN modulates DNA replication by direct binding to Cdt1, and thereby alters its stability and activity. GMNN is involved in various stages of development such as pre-implantation, germ layer formation, cell commitment and specification, maintenance of genome integrity at mid blastula transition, epithelial to mesenchymal transition during gastrulation, neural development, organogenesis and axis patterning. GMNN interacts with different proteins resulting in enhanced hematopoietic stem cell activity thereby activating the development-associated genes' transcription. GMNN expression is also associated with cancer pathophysiology and development. In this review we discussed the structure and function of GMNN in detail. Inhibitors of GMNN and their role in DNA replication, repair, cell cycle and apoptosis are reviewed. Further, we also discussed the role of GMNN in virus infected host cells.


Asunto(s)
Ciclo Celular/genética , Replicación del ADN/genética , Embrión no Mamífero/metabolismo , Geminina/genética , Neoplasias/genética , Animales , Embrión no Mamífero/embriología , Geminina/química , Geminina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Neoplasias/fisiopatología , Conformación Proteica
6.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2278-86, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26527144

RESUMEN

GemC1, together with Idas and Geminin, an important regulator of DNA-replication licensing and differentiation decisions, constitute a superfamily sharing a homologous central coiled-coil domain. To better understand this family of proteins, the crystal structure of a GemC1 coiled-coil domain variant engineered for better solubility was determined to 2.2 Å resolution. GemC1 shows a less typical coiled coil compared with the Geminin homodimer and the Geminin-Idas heterodimer structures. It is also shown that both in vitro and in cells GemC1 interacts with Geminin through its coiled-coil domain, forming a heterodimer that is more stable that the GemC1 homodimer. Comparative analysis of the thermal stability of all of the possible superfamily complexes, using circular dichroism to follow the unfolding of the entire helix of the coiled coil, or intrinsic tryptophan fluorescence of a unique conserved N-terminal tryptophan, shows that the unfolding of the coiled coil is likely to take place from the C-terminus towards the N-terminus. It is also shown that homodimers show a single-state unfolding, while heterodimers show a two-state unfolding, suggesting that the dimer first falls apart and the helices then unfold according to the stability of each protein. The findings argue that Geminin-family members form homodimers and heterodimers between them, and this ability is likely to be important for modulating their function in cycling and differentiating cells.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Geminina/metabolismo , Secuencia de Aminoácidos , Proteínas de Ciclo Celular , Cristalografía por Rayos X , Geminina/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Mapas de Interacción de Proteínas , Multimerización de Proteína , Estabilidad Proteica , Temperatura
7.
J Biol Chem ; 289(44): 30810-30821, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25231993

RESUMEN

The initial step in initiation of eukaryotic DNA replication involves the assembly of pre-replicative complexes (pre-RCs) at origins of replication during the G1 phase of the cell cycle. In metazoans initiation is inhibited by the regulatory factor Geminin. We have purified the human pre-RC proteins, studied their interactions in vitro with each other and with origin DNA, and analyzed the effects of HsGeminin on formation of DNA-protein complexes. The formation of an initial complex containing the human origin recognition complex (HsORC), HsCdt1, HsCdc6, and origin DNA is cooperative, involving all possible binary interactions among the components. Maximal association of HsMCM2-7, a component of the replicative helicase, requires HsORC, HsCdc6, HsCdt1, and ATP, and is driven by interactions of HsCdt1 and HsCdc6 with multiple HsMCM2-7 subunits. Formation of stable complexes, resistant to high salt, requires ATP hydrolysis. In the absence of HsMCM proteins, HsGeminin inhibits the association of HsCdt1 with DNA or with HsORC-HsCdc6-DNA complexes. However, HsGeminin does not inhibit recruitment of HsMCM2-7 to DNA to form complexes containing all of the pre-RC proteins. In fact, HsGeminin itself is a component of such complexes, and interacts directly with the HsMcm3 and HsMcm5 subunits of HsMCM2-7, as well as with HsCdt1. Although HsGeminin does not prevent the initial formation of DNA-protein complexes containing the pre-RC proteins, it strongly inhibits the formation of stable pre-RCs that are resistant to high salt. We suggest that bound HsGeminin prevents transition of the pre-RC to a state that is competent for initiation of DNA replication.


Asunto(s)
Replicación del ADN , Geminina/química , Ácidos Nucleicos Inmovilizados/química , Proteínas de Ciclo Celular/química , Células HEK293 , Humanos , Proteínas de Mantenimiento de Minicromosoma/química , Proteínas Nucleares/química , Complejo de Reconocimiento del Origen/química , Unión Proteica , Estabilidad Proteica
8.
J Biol Chem ; 288(44): 31624-34, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24064211

RESUMEN

Geminin is an important regulator of proliferation and differentiation in metazoans, which predominantly inhibits the DNA replication licensing factor Cdt1, preventing genome over-replication. We show that Geminin preferentially forms stable coiled-coil heterodimers with its homologue, Idas. In contrast to Idas-Geminin heterodimers, Idas homodimers are thermodynamically unstable and are unlikely to exist as a stable macromolecule under physiological conditions. The crystal structure of the homology regions of Idas in complex with Geminin showed a tight head-to-head heterodimeric coiled-coil. This Idas-Geminin heterodimer binds Cdt1 less strongly than Geminin-Geminin, still with high affinity (∼30 nm), but with notably different thermodynamic properties. Consistently, in Xenopus egg extracts, Idas-Geminin is less active in licensing inhibition compared with a Geminin-Geminin homodimer. In human cultured cells, ectopic expression of Idas leads to limited over-replication, which is counteracted by Geminin co-expression. The properties of the Idas-Geminin complex suggest it as the functional form of Idas and provide a possible mechanism to modulate Geminin activity.


Asunto(s)
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN/fisiología , Geminina/química , Geminina/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Multimerización de Proteína/fisiología , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Geminina/genética , Humanos , Proteínas Nucleares/genética , Estructura Cuaternaria de Proteína , Relación Estructura-Actividad , Factores de Transcripción , Xenopus laevis
9.
Nat Commun ; 4: 1885, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23695679

RESUMEN

Geminin, an essential factor for DNA replication, directly binds to the licensing factor Cdt1 and inhibits pre-replicative complex formation to prevent re-replication. In G1, geminin levels are controlled by the anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase complex, which targets geminin for proteasomal degradation to allow pre-replicative complex formation. Conversely, from S to G2, geminin is stabilized due to APC/C ubiquitin ligase complex inhibition, ensuring the inhibition of pre-replicative complex formation. However, mitotic regulation of geminin has hitherto not been described. Here we show that Aurora-A phosphorylates geminin on Thr25 during M phase, and this event induces geminin stabilization by preventing its APC/C ubiquitin ligase complex-mediated degradation during mitosis. In turn, stabilized geminin inhibits SCF(Skp2)-mediated degradation of Cdt1 to ensure pre-replicative complex formation in the ensuing S phase. The Aurora-A-geminin-Cdt1 axis therefore represents a critical regulator of proper DNA replication.


Asunto(s)
Aurora Quinasa A/metabolismo , Replicación del ADN , Geminina/metabolismo , Mitosis , Secuencia de Aminoácidos , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Aurora Quinasa A/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Regulación hacia Abajo , Geminina/química , Humanos , Datos de Secuencia Molecular , Fosforilación , Fosfotreonina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Solubilidad
10.
Subcell Biochem ; 62: 71-87, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22918581

RESUMEN

One of the mechanisms controlling the initiation of DNA replication is the dynamic interaction between Cdt1, which promotes assembly of the pre-replication license complex, and Geminin, which inhibits it. Specifically, Cdt1 cooperates with the cell cycle protein Cdc6 to promote loading of the minichromosome maintenance helicases (MCM) onto the chromatin-bound origin recognition complex (ORC), by directly interacting with the MCM complex, and by modulating histone acetylation and inducing chromatin unfolding. Geminin, on the other hand, prevents the loading of the MCM onto the ORC both by directly binding to Cdt1, and by modulating Cdt1 stability and activity. Protein levels of Geminin and Cdt1 are tightly regulated through the cell cycle, and the Cdt1-Geminin complex likely acts as a molecular switch that can enable or disable the firing of each origin of replication. In this review we summarize structural studies of Cdt1 and Geminin and subsequent insights into how this molecular switch may function to ensure DNA is faithfully replicated only once during S phase of each cell cycle.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Geminina/metabolismo , Complejo de Reconocimiento del Origen/metabolismo , Fase S/fisiología , Acetilación , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Geminina/química , Geminina/genética , Histonas/química , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo de Reconocimiento del Origen/química , Complejo de Reconocimiento del Origen/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA