Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell Biol ; 43(5): 200-222, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37114947

RESUMEN

Rho GTPases are global regulators of cell polarity and signaling. By exploring the turnover regulation of the yeast Rho GTPase Cdc42p, we identified new regulatory features surrounding the stability of the protein. We specifically show that Cdc42p is degraded at 37 °C by chaperones through lysine residues located in the C-terminus of the protein. Cdc42p turnover at 37 °C occurred by the 26S proteasome in an ESCRT-dependent manner in the lysosome/vacuole. By analyzing versions of Cdc42p that were defective for turnover, we show that turnover at 37 °C promoted cell polarity but was defective for sensitivity to mating pheromone, presumably mediated through a Cdc42p-dependent MAP kinase pathway. We also identified one residue (K16) in the P-loop of the protein that was critical for Cdc42p stability. Accumulation of Cdc42pK16R in some contexts led to the formation of protein aggregates, which were enriched in aging mother cells and cells undergoing proteostatic stress. Our study uncovers new aspects of protein turnover regulation of a Rho-type GTPase that may extend to other systems. Moreover, residues identified here that mediate Cdc42p turnover correlate with several human diseases, which may suggest that turnover regulation of Cdc42p is important to aspects of human health.


Asunto(s)
Polaridad Celular , Proteínas de Saccharomyces cerevisiae , Proteína de Unión al GTP cdc42 de Saccharomyces cerevisiae , Humanos , Proteína de Unión al GTP cdc42 de Saccharomyces cerevisiae/metabolismo , Polaridad Celular/fisiología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal
2.
Sci Adv ; 8(18): eabm4086, 2022 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-35522754

RESUMEN

Cells maintain their size within limits over successive generations to maximize fitness and survival. Sizer, timer, and adder behaviors have been proposed as possible alternatives to coordinate growth and cell cycle progression. Regarding budding yeast cells, a sizer mechanism is thought to rule cell cycle entry at Start. However, while many proteins controlling the size of these cells have been identified, the mechanistic framework in which they participate to achieve cell size homeostasis is not understood. We show here that intertwined APC and SCF degradation machineries with specific adaptor proteins drive cyclic accumulation of the G1 Cdk in the nucleus, reaching maximal levels at Start. The mechanism incorporates Mad3, a centromeric-signaling protein that subordinates G1 progression to the previous mitosis as a memory factor. This alternating-degradation device displays the properties of a timer and, together with the sizer device, would constitute a key determinant of cell cycle entry.

4.
J Cell Biol ; 220(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33480968

RESUMEN

Stress granules (SGs) are conserved biomolecular condensates that originate in response to many stress conditions. These membraneless organelles contain nontranslating mRNAs and a diverse subproteome, but our knowledge of their regulation and functional relevance is still incipient. Here, we describe a mutual-inhibition interplay between SGs and Cdc28, the budding yeast Cdk. Among Cdc28 interactors acting as negative modulators of Start, we have identified Whi8, an RNA-binding protein that localizes to SGs and recruits the mRNA of CLN3, the most upstream G1 cyclin, for efficient translation inhibition and Cdk inactivation under stress. However, Whi8 also contributes to recruiting Cdc28 to SGs, where it acts to promote their dissolution. As predicted by a mutual-inhibition framework, the SG constitutes a bistable system that is modulated by Cdk. Since mammalian cells display a homologous mechanism, we propose that the opposing functions of specific mRNA-binding proteins and Cdk's subjugate SG dynamics to a conserved hysteretic switch.


Asunto(s)
Proteína Quinasa CDC28 de Saccharomyces cerevisiae/metabolismo , Gránulos Citoplasmáticos/metabolismo , Saccharomyces cerevisiae/metabolismo , Estrés Fisiológico , Ciclo Celular , Ciclinas/metabolismo , Células HeLa , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Modelos Biológicos , Unión Proteica , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
5.
Exp Cell Res ; 394(2): 112163, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32640194

RESUMEN

Aging is characterized by the progressive decline of physiology at the cell, tissue and organism level, leading to an increased risk of mortality. Proteotoxic stress, mitochondrial dysfunction and genomic instability are considered major universal drivers of cell aging, and accumulating evidence establishes clear biunivocal relationships among these key hallmarks. In this regard, the finite lifespan of the budding yeast, together with the extensive armamentarium of available analytical tools, has made this single cell eukaryote a key model to study aging at molecular and cellular levels. Here we review the current data that link proteostasis to cell cycle progression in the budding yeast, focusing on senescence as an inherent phenotype displayed by aged cells. Recent advances in high-throughput systems to study yeast mother cells while they replicate are providing crucial information on aging-related processes and their temporal interdependencies at a systems level. In our view, the available data point to the existence of multiple feedback mechanisms among the major causal factors of aging, which would converge into the loss of proteostasis as a nodal driver of cell senescence and death.


Asunto(s)
Senescencia Celular , Replicación del ADN , Proteostasis , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Ciclo Celular , Retroalimentación Fisiológica
6.
Elife ; 82019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31518229

RESUMEN

Loss of proteostasis and cellular senescence are key hallmarks of aging, but direct cause-effect relationships are not well understood. We show that most yeast cells arrest in G1 before death with low nuclear levels of Cln3, a key G1 cyclin extremely sensitive to chaperone status. Chaperone availability is seriously compromised in aged cells, and the G1 arrest coincides with massive aggregation of a metastable chaperone-activity reporter. Moreover, G1-cyclin overexpression increases lifespan in a chaperone-dependent manner. As a key prediction of a model integrating autocatalytic protein aggregation and a minimal Start network, enforced protein aggregation causes a severe reduction in lifespan, an effect that is greatly alleviated by increased expression of specific chaperones or cyclin Cln3. Overall, our data show that proteostasis breakdown, by compromising chaperone activity and G1-cyclin function, causes an irreversible arrest in G1, configuring a molecular pathway postulating proteostasis decay as a key contributing effector of cell senescence.


Asunto(s)
Puntos de Control del Ciclo Celular , Senescencia Celular , Chaperonas Moleculares/metabolismo , Proteostasis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/crecimiento & desarrollo , Ciclinas/metabolismo
7.
Methods Mol Biol ; 2040: 375-384, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31432488

RESUMEN

The dynamics of cellular processes is a crucial aspect to consider when trying to understand cell function, particularly with regard to the coordination of complex mechanisms involving extensive molecular networks in different cell compartments. Thus, there is an urgent demand of methodologies able to obtain accurate spatiotemporal information on molecular dynamics in live cells. Different variants based on fluorescence correlation spectroscopy have been used successfully in the analysis of protein diffusion and complex or aggregation status. However, the available approaches are limited when simultaneous spatial and temporal resolutions are required to analyze fast processes. Here we describe the use of raster image correlation spectroscopy to analyze the spatiotemporal coincidence of collaborating proteins in highly dynamic molecular mechanisms.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Microscopía Intravital/métodos , Imagen de Lapso de Tiempo/métodos , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/genética , Microscopía Intravital/instrumentación , Sustancias Luminiscentes/química , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Análisis Espacio-Temporal , Espectrometría de Fluorescencia/métodos , Imagen de Lapso de Tiempo/instrumentación , Proteína Fluorescente Roja
8.
Life Sci Alliance ; 2(2)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30988162

RESUMEN

The precise coordination of growth and proliferation has a universal prevalence in cell homeostasis. As a prominent property, cell size is modulated by the coordination between these processes in bacterial, yeast, and mammalian cells, but the underlying molecular mechanisms are largely unknown. Here, we show that multifunctional chaperone systems play a concerted and limiting role in cell-cycle entry, specifically driving nuclear accumulation of the G1 Cdk-cyclin complex. Based on these findings, we establish and test a molecular competition model that recapitulates cell-cycle-entry dependence on growth rate. As key predictions at a single-cell level, we show that availability of the Ydj1 chaperone and nuclear accumulation of the G1 cyclin Cln3 are inversely dependent on growth rate and readily respond to changes in protein synthesis and stress conditions that alter protein folding requirements. Thus, chaperone workload would subordinate Start to the biosynthetic machinery and dynamically adjust proliferation to the growth potential of the cell.


Asunto(s)
Aumento de la Célula , Tamaño de la Célula , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Respuesta al Choque Térmico/fisiología , Chaperonas Moleculares/metabolismo , Estrés Salino/fisiología , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/metabolismo , Nucléolo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Modelos Moleculares , Puntos de Control de la Fase S del Ciclo Celular/fisiología , Saccharomyces cerevisiae/citología , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
PLoS Biol ; 16(8): e2005388, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30080861

RESUMEN

Cell size scales with ploidy in a great range of eukaryotes, but the underlying mechanisms remain unknown. Using various orthogonal single-cell approaches, we show that cell size increases linearly with centromere (CEN) copy number in budding yeast. This effect is due to a G1 delay mediated by increased degradation of Cln3, the most upstream G1 cyclin acting at Start, and specific centromeric signaling proteins, namely Mad3 and Bub3. Mad3 binds both Cln3 and Cdc4, the adaptor component of the Skp1/Cul1/F-box (SCF) complex that targets Cln3 for degradation, these interactions being essential for the CEN-dosage dependent effects on cell size. Our results reveal a pathway that modulates cell size as a function of CEN number, and we speculate that, in cooperation with other CEN-independent mechanisms, it could assist the cell to attain efficient mass/ploidy ratios.


Asunto(s)
Procesos de Crecimiento Celular/fisiología , Centrómero/fisiología , Ciclina G1/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular , Aumento de la Célula , Centrómero/metabolismo , Ciclinas/metabolismo , Fase G1/fisiología , Regulación Fúngica de la Expresión Génica , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiología , Proteínas Nucleares/metabolismo , Proteolisis , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomycetales/metabolismo , Transducción de Señal
10.
EMBO J ; 37(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29950310

RESUMEN

Cells sense myriad signals during G1, and a rapid response to prevent cell cycle entry is of crucial importance for proper development and adaptation. Cln3, the most upstream G1 cyclin in budding yeast, is an extremely short-lived protein subject to ubiquitination and proteasomal degradation. On the other hand, nuclear accumulation of Cln3 depends on chaperones that are also important for its degradation. However, how these processes are intertwined to control G1-cyclin fate is not well understood. Here, we show that Cln3 undergoes a challenging ubiquitination step required for both degradation and full activation. Segregase Cdc48/p97 prevents degradation of ubiquitinated Cln3, and concurrently stimulates its ER release and nuclear accumulation to trigger Start. Cdc48/p97 phosphorylation at conserved Cdk-target sites is important for recruitment of specific cofactors and, in both yeast and mammalian cells, to attain proper G1-cyclin levels and activity. Cdk-dependent modulation of Cdc48 would subjugate G1 cyclins to fast and reversible state switching, thus arresting cells promptly in G1 at developmental or environmental checkpoints, but also resuming G1 progression immediately after proliferative signals reappear.


Asunto(s)
Ciclinas/metabolismo , Fase G1/fisiología , Proteolisis , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteína que Contiene Valosina/metabolismo , Células 3T3 , Animales , Ciclinas/genética , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Proteína que Contiene Valosina/genética
11.
Front Cell Dev Biol ; 5: 57, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28603712

RESUMEN

Cells are able to adjust their growth and size to external inputs to comply with specific fates and developmental programs. Molecular pathways controlling growth also have an enormous impact in cell size, and bacteria, yeast, or epithelial cells modify their size as a function of growth rate. This universal feature suggests that growth (mass) and proliferation (cell number) rates are subject to general coordinating mechanisms. However, the underlying molecular connections are still a matter of debate. Here we review the current ideas on growth and cell size control, and focus on the possible mechanisms that could link the biosynthetic machinery to the Start network in budding yeast. In particular, we discuss the role of molecular chaperones in a competition framework to explain cell size control by growth at the individual cell level.

12.
Curr Biol ; 27(6): 773-783, 2017 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-28262489

RESUMEN

In order to produce rejuvenated daughters, dividing budding yeast cells confine aging factors, including protein aggregates, to the aging mother cell. The asymmetric inheritance of these protein deposits is mediated by organelle and cytoskeletal attachment and by cell geometry. Yet it remains unclear how deposit formation is restricted to the aging lineage. Here, we show that selective membrane anchoring and the compartmentalization of the endoplasmic reticulum (ER) membrane confine protein deposit formation to aging cells during division. Supporting the idea that the age-dependent deposit forms through coalescence of smaller aggregates, two deposits rapidly merged when placed in the same cell by cell-cell fusion. The deposits localized to the ER membrane, primarily to the nuclear envelope (NE). Strikingly, weakening the diffusion barriers that separate the ER membrane into mother and bud compartments caused premature formation of deposits in the daughter cells. Detachment of the Hsp40 protein Ydj1 from the ER membrane elicited a similar phenotype, suggesting that the diffusion barriers and farnesylated Ydj1 functioned together to confine protein deposit formation to mother cells during division. Accordingly, fluorescence correlation spectroscopy measurements in dividing cells indicated that a slow-diffusing, possibly client-bound Ydj1 fraction was asymmetrically enriched in the mother compartment. This asymmetric distribution depended on Ydj1 farnesylation and intact diffusion barriers. Taking these findings together, we propose that ER-anchored Ydj1 binds deposit precursors and prevents them from spreading into daughter cells during division by subjecting them to the ER diffusion barriers. This ensures that the coalescence of precursors into a single deposit is restricted to the aging lineage.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Biosíntesis de Proteínas , Saccharomyces cerevisiae/genética
13.
Sci Rep ; 6: 19729, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26818620

RESUMEN

Nucleosomes provide additional regulatory mechanisms to transcription and DNA replication by mediating the access of proteins to DNA. During the cell cycle chromatin undergoes several conformational changes, however the functional significance of these changes to cellular processes are largely unexplored. Here, we present the first comprehensive genome-wide study of nucleosome plasticity at single base-pair resolution along the cell cycle in Saccharomyces cerevisiae. We determined nucleosome organization with a specific focus on two regulatory regions: transcription start sites (TSSs) and replication origins (ORIs). During the cell cycle, nucleosomes around TSSs display rearrangements in a cyclic manner. In contrast to gap (G1 and G2) phases, nucleosomes have a fuzzier organization during S and M phases, Moreover, the choreography of nucleosome rearrangements correlate with changes in gene expression during the cell cycle, indicating a strong association between nucleosomes and cell cycle-dependent gene functionality. On the other hand, nucleosomes are more dynamic around ORIs along the cell cycle, albeit with tighter regulation in early firing origins, implying the functional role of nucleosomes on replication origins. Our study provides a dynamic picture of nucleosome organization throughout the cell cycle and highlights the subsequent impact on transcription and replication activity.


Asunto(s)
Ciclo Celular/fisiología , Replicación del ADN/fisiología , ADN de Hongos/biosíntesis , Nucleosomas/metabolismo , Saccharomyces cerevisiae/metabolismo , ADN de Hongos/genética , Nucleosomas/genética , Saccharomyces cerevisiae/genética
14.
Nat Methods ; 12(10): 955-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26322837

RESUMEN

Protein tagging is widely used in approaches ranging from affinity purification to fluorescence-based detection in live cells. However, an intrinsic limitation of tagging is that the native function of the protein may be compromised or even abolished by the presence of the tag. Here we describe and characterize a set of small, innocuous protein tags (inntags) that we anticipate will find application in a variety of biological techniques.


Asunto(s)
Epítopos/análisis , Epítopos/química , Técnica del Anticuerpo Fluorescente/métodos , Inmunoprecipitación/métodos , Proteínas/análisis , Proteínas/inmunología , Animales , Anticuerpos Monoclonales , Epítopos/genética , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
15.
J Neurosci ; 34(42): 13988-97, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25319695

RESUMEN

Local regulation of protein synthesis allows a neuron to rapidly alter the proteome in response to synaptic signals, an essential mechanism in synaptic plasticity that is altered in many neurological diseases. Synthesis of many synaptic proteins is under local control and much of this regulation occurs through structures termed RNA granules. KIS is a protein kinase that associates with stathmin, a modulator of the tubulin cytoskeleton. Furthermore, KIS is found in RNA granules and stimulates translation driven by the ß-actin 3'UTR in neurites. Here we explore the physiological and molecular mechanisms underlying the action of KIS on hippocampal synaptic plasticity in mice. KIS downregulation compromises spine development, alters actin dynamics, and reduces postsynaptic responsiveness. The absence of KIS results in a significant decrease of protein levels of PSD-95, a postsynaptic scaffolding protein, and the AMPAR subunits GluR1 and GluR2 in a CPEB3-dependent manner. Underlying its role in spine maturation, KIS is able to suppress the spine developmental defects caused by CPEB3 overexpression. Moreover, either by direct or indirect mechanisms, KIS counteracts the inhibitory activity of CPEB3 on the GluR2 3'UTR at both mRNA translation and polyadenylation levels. Our study provides insights into the mechanisms that mediate dendritic spine morphogenesis and functional synaptic maturation, and suggests KIS as a link regulating spine cytoskeleton and postsynaptic activity in memory formation.


Asunto(s)
Espinas Dendríticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Microtúbulos/fisiología , Plasticidad Neuronal/fisiología , Biosíntesis de Proteínas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores AMPA/biosíntesis , Animales , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Técnicas de Cultivo de Órganos
16.
Mol Cell ; 53(1): 115-26, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24374311

RESUMEN

Cells commit to a new cell cycle at Start by activation of the G1 Cdk-cyclin complex which, in turn, triggers a genome-wide transcriptional wave that executes the G1/S transition. In budding yeast, the Cdc28-Cln3 complex is regulated by an ER-retention mechanism that is important for proper cell size control. We have isolated small-cell-size CDC28 mutants showing impaired retention at the ER and premature accumulation of the Cln3 cyclin in the nucleus. The differential interactome of a quintuple Cdc28(wee) mutant pinpointed Whi7, a Whi5 paralog targeted by Cdc28 that associates to the ER in a phosphorylation-dependent manner. Our results demonstrate that the Cln3 cyclin and Whi7 act in a positive feedback loop to release the G1 Cdk-cyclin complex and trigger Start once a critical size has been reached, thus uncovering a key nonlinear mechanism at the earliest known events of cell-cycle entry.


Asunto(s)
Proteína Quinasa CDC28 de Saccharomyces cerevisiae/metabolismo , Ciclinas/metabolismo , Retículo Endoplásmico/metabolismo , Fase G1/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/genética , Ciclinas/genética , Retículo Endoplásmico/genética , Mutación , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
17.
Nat Commun ; 3: 1012, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22910358

RESUMEN

Budding yeast cells are assumed to trigger Start and enter the cell cycle only after they attain a critical size set by external conditions. However, arguing against deterministic models of cell size control, cell volume at Start displays great individual variability even under constant conditions. Here we show that cell size at Start is robustly set at a single-cell level by the volume growth rate in G1, which explains the observed variability. We find that this growth-rate-dependent sizer is intimately hardwired into the Start network and the Ydj1 chaperone is key for setting cell size as a function of the individual growth rate. Mathematical modelling and experimental data indicate that a growth-rate-dependent sizer is sufficient to ensure size homeostasis and, as a remarkable advantage over a rigid sizer mechanism, it reduces noise in G1 length and provides an immediate solution for size adaptation to external conditions at a population level.


Asunto(s)
Ciclo Celular , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/crecimiento & desarrollo , Fase G1 , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Homeostasis , Cinética , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
18.
Traffic ; 12(5): 549-62, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21306487

RESUMEN

Nuclear accumulation of cyclin D1 because of altered trafficking or degradation is thought to contribute directly to neoplastic transformation and growth. Mechanisms of cyclin D1 localization in S phase have been studied in detail, but its control during exit from the cell cycle and quiescence is poorly understood. Here we report that translokin (Tlk), a microtubule-associated protein also termed Cep57, interacts with cyclin D1 and controls its nucleocytoplasmic distribution in quiescent cells. Tlk binds to regions of cyclin D1 also involved in binding to cyclin-dependent kinase 4 (Cdk4), and a fraction of cyclin D1 associates to the juxtanuclear Tlk network in the cell. Downregulation of Tlk levels results in undue nuclear accumulation of cyclin D1 and increased Cdk4-dependent phosphorylation of pRB under quiescence conditions. In turn, overexpression of Tlk prevents proper cyclin D1 accumulation in the nucleus of proliferating cells in an interaction-dependent manner, inhibits Cdk4-dependent phosphorylation of pRB and hinders cell cycle progression to S phase. We propose that the Tlk acts as a key negative regulator in the pathway that drives nuclear import of cyclin D1, thus contributing to prevent pRB inactivation and to maintain cellular quiescence.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclo Celular/fisiología , Núcleo Celular/metabolismo , Ciclina D1/metabolismo , Fibroblastos/metabolismo , Animales , Proteínas de Ciclo Celular , Células Cultivadas , Quinasa 4 Dependiente de la Ciclina/metabolismo , Fibroblastos/citología , Humanos , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/metabolismo
19.
Genome Biol ; 11(6): R67, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20573214

RESUMEN

BACKGROUND: The G1-to-S transition of the cell cycle in the yeast Saccharomyces cerevisiae involves an extensive transcriptional program driven by transcription factors SBF (Swi4-Swi6) and MBF (Mbp1-Swi6). Activation of these factors ultimately depends on the G1 cyclin Cln3. RESULTS: To determine the transcriptional targets of Cln3 and their dependence on SBF or MBF, we first have used DNA microarrays to interrogate gene expression upon Cln3 overexpression in synchronized cultures of strains lacking components of SBF and/or MBF. Secondly, we have integrated this expression dataset together with other heterogeneous data sources into a single probabilistic model based on Bayesian statistics. Our analysis has produced more than 200 transcription factor-target assignments, validated by ChIP assays and by functional enrichment. Our predictions show higher internal coherence and predictive power than previous classifications. Our results support a model whereby SBF and MBF may be differentially activated by Cln3. CONCLUSIONS: Integration of heterogeneous genome-wide datasets is key to building accurate transcriptional networks. By such integration, we provide here a reliable transcriptional network at the G1-to-S transition in the budding yeast cell cycle. Our results suggest that to improve the reliability of predictions we need to feed our models with more informative experimental data.


Asunto(s)
Ciclinas/metabolismo , Fase G1/genética , Regulación Fúngica de la Expresión Génica , Redes Reguladoras de Genes/genética , Fase S/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Inmunoprecipitación de Cromatina , Ciclinas/genética , ADN de Hongos/metabolismo , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Genes Fúngicos/genética , Variación Genética , Funciones de Verosimilitud , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Unión Proteica , Reproducibilidad de los Resultados , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Transcripción Genética
20.
J Biol Chem ; 284(47): 32980-8, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19801649

RESUMEN

E47 is a basic helix-loop-helix transcription factor involved in neuronal differentiation and survival. We had previously shown that the basic helix-loop-helix protein E47 binds to E-box sequences within the promoter of the TrkB gene and activates its transcription. Proper expression of the TrkB receptor plays a key role in development and function of the vertebrate nervous system, and altered levels of TrkB have been associated with important human diseases. Here we show that E47 interacts with MLK2, a mixed lineage kinase (MLK) involved in JNK-mediated activation of programmed cell death. MLK2 enhances phosphorylation of the AD2 activation domain of E47 in vivo in a JNK-independent manner and phosphorylates in vitro defined serine and threonine residues within a loop-helix structure of AD2 that also contains a putative MLK docking site. Although these residues are essential for MLK2-mediated inactivation of E47, inhibition of MLKs by CEP11004 causes up-regulation of TrkB at a transcriptional level in cerebellar granule neurons and differentiating neuroblastoma cells. These findings allow us to propose a novel mechanism by which MLK regulates TrkB expression through phosphorylation of an activation domain of E47. This molecular link would explain why MLK inhibitors not only prevent activation of cell death processes but also enhance cell survival signaling as a key aspect of their neuroprotective potential.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Quinasas Quinasa Quinasa PAM/metabolismo , Neuronas/metabolismo , Receptor trkB/biosíntesis , Factores de Transcripción TCF/fisiología , Animales , Muerte Celular , Línea Celular Tumoral , Supervivencia Celular , Dimerización , Humanos , Ratones , Fosforilación , Factores de Transcripción TCF/metabolismo , Proteína 1 Similar al Factor de Transcripción 7 , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...