Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.617
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 172(5): 1007-1021.e17, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474905

RESUMEN

MLL/SET methyltransferases catalyze methylation of histone 3 lysine 4 and play critical roles in development and cancer. We assessed MLL/SET proteins and found that SETD1A is required for survival of acute myeloid leukemia (AML) cells. Mutagenesis studies and CRISPR-Cas9 domain screening show the enzymatic SET domain is not necessary for AML cell survival but that a newly identified region termed the "FLOS" (functional location on SETD1A) domain is indispensable. FLOS disruption suppresses DNA damage response genes and induces p53-dependent apoptosis. The FLOS domain acts as a cyclin-K-binding site that is required for chromosomal recruitment of cyclin K and for DNA-repair-associated gene expression in S phase. These data identify a connection between the chromatin regulator SETD1A and the DNA damage response that is independent of histone methylation and suggests that targeting SETD1A and cyclin K complexes may represent a therapeutic opportunity for AML and, potentially, for other cancers.


Asunto(s)
Ciclinas/metabolismo , Daño del ADN , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Biocatálisis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Ciclinas/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Histonas , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Unión Proteica , Dominios Proteicos , Estabilidad Proteica , Transcripción Genética
2.
Mol Cell ; 84(7): 1224-1242.e13, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38458201

RESUMEN

Although mismatch repair (MMR) is essential for correcting DNA replication errors, it can also recognize other lesions, such as oxidized bases. In G0 and G1, MMR is kept in check through unknown mechanisms as it is error-prone during these cell cycle phases. We show that in mammalian cells, D-type cyclins are recruited to sites of oxidative DNA damage in a PCNA- and p21-dependent manner. D-type cyclins inhibit the proteasomal degradation of p21, which competes with MMR proteins for binding to PCNA, thereby inhibiting MMR. The ability of D-type cyclins to limit MMR is CDK4- and CDK6-independent and is conserved in G0 and G1. At the G1/S transition, the timely, cullin-RING ubiquitin ligase (CRL)-dependent degradation of D-type cyclins and p21 enables MMR activity to efficiently repair DNA replication errors. Persistent expression of D-type cyclins during S-phase inhibits the binding of MMR proteins to PCNA, increases the mutational burden, and promotes microsatellite instability.


Asunto(s)
Ciclinas , Reparación de la Incompatibilidad de ADN , Animales , Ciclinas/genética , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Interfase , Mamíferos/metabolismo
3.
Mol Cell ; 84(11): 2119-2134.e5, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38848691

RESUMEN

Protein synthesis is metabolically costly and must be tightly coordinated with changing cellular needs and nutrient availability. The cap-binding protein eIF4E makes the earliest contact between mRNAs and the translation machinery, offering a key regulatory nexus. We acutely depleted this essential protein and found surprisingly modest effects on cell growth and recovery of protein synthesis. Paradoxically, impaired protein biosynthesis upregulated genes involved in the catabolism of aromatic amino acids simultaneously with the induction of the amino acid biosynthetic regulon driven by the integrated stress response factor GCN4. We further identified the translational control of Pho85 cyclin 5 (PCL5), a negative regulator of Gcn4, that provides a consistent protein-to-mRNA ratio under varied translation environments. This regulation depended in part on a uniquely long poly(A) tract in the PCL5 5' UTR and poly(A) binding protein. Collectively, these results highlight how eIF4E connects protein synthesis to metabolic gene regulation, uncovering mechanisms controlling translation during environmental challenges.


Asunto(s)
Aminoácidos , Factor 4E Eucariótico de Iniciación , Regulación Fúngica de la Expresión Génica , Biosíntesis de Proteínas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Factor 4E Eucariótico de Iniciación/metabolismo , Factor 4E Eucariótico de Iniciación/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Aminoácidos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , ARN Mensajero/metabolismo , ARN Mensajero/genética , Regiones no Traducidas 5' , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Unión a Poli(A)/metabolismo , Proteínas de Unión a Poli(A)/genética
4.
EMBO J ; 43(11): 2094-2126, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38600241

RESUMEN

A versatile division of apicomplexan parasites and a dearth of conserved regulators have hindered the progress of apicomplexan cell cycle studies. While most apicomplexans divide in a multinuclear fashion, Toxoplasma gondii tachyzoites divide in the traditional binary mode. We previously identified five Toxoplasma CDK-related kinases (Crk). Here, we investigated TgCrk4 and its cyclin partner TgCyc4. We demonstrated that TgCrk4 regulates conventional G2 phase processes, such as repression of chromosome rereplication and centrosome reduplication, and acts upstream of the spindle assembly checkpoint. The spatial TgCyc4 dynamics supported the TgCrk4-TgCyc4 complex role in the coordination of chromosome and centrosome cycles. We also identified a dominant TgCrk4-TgCyc4 complex interactor, TgiRD1 protein, related to DNA replication licensing factor CDT1 but played no role in licensing DNA replication in the G1 phase. Our results showed that TgiRD1 also plays a role in controlling chromosome and centrosome reduplication. Global phosphoproteome analyses identified TgCrk4 substrates, including TgORC4, TgCdc20, TgGCP2, and TgPP2ACA. Importantly, the phylogenetic and structural studies suggest the Crk4-Cyc4 complex is limited to a minor group of the binary dividing apicomplexans.


Asunto(s)
Proteínas Protozoarias , Toxoplasma , Toxoplasma/metabolismo , Toxoplasma/genética , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Fase G2/genética , Centrosoma/metabolismo , División Celular , Ciclinas/metabolismo , Ciclinas/genética
5.
Mol Cell ; 78(2): 359-370.e6, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32246903

RESUMEN

Yeast cells must grow to a critical size before committing to division. It is unknown how size is measured. We find that as cells grow, mRNAs for some cell-cycle activators scale faster than size, increasing in concentration, while mRNAs for some inhibitors scale slower than size, decreasing in concentration. Size-scaled gene expression could cause an increasing ratio of activators to inhibitors with size, triggering cell-cycle entry. Consistent with this, expression of the CLN2 activator from the promoter of the WHI5 inhibitor, or vice versa, interfered with cell size homeostasis, yielding a broader distribution of cell sizes. We suggest that size homeostasis comes from differential scaling of gene expression with size. Differential regulation of gene expression as a function of cell size could affect many cellular processes.


Asunto(s)
División Celular/genética , Tamaño de la Célula , Ciclinas/genética , Proteínas de Saccharomyces cerevisiae/genética , Ciclo Celular/genética , Fase G1/genética , Regulación del Desarrollo de la Expresión Génica/genética , Regulación Fúngica de la Expresión Génica/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo
6.
EMBO J ; 42(24): e114051, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-38059508

RESUMEN

CDK11 is an emerging druggable target for cancer therapy due to its prevalent roles in phosphorylating critical transcription and splicing factors and in facilitating cell cycle progression in cancer cells. Like other cyclin-dependent kinases, CDK11 requires its cognate cyclin, cyclin L1 or cyclin L2, for activation. However, little is known about how CDK11 activities might be modulated by other regulators. In this study, we show that CDK11 forms a tight complex with cyclins L1/L2 and SAP30BP, the latter of which is a poorly characterized factor. Acute degradation of SAP30BP mirrors that of CDK11 in causing widespread and strong defects in pre-mRNA splicing. Furthermore, we demonstrate that SAP30BP facilitates CDK11 kinase activities in vitro and in vivo, through ensuring the stabilities and the assembly of cyclins L1/L2 with CDK11. Together, these findings uncover SAP30BP as a critical CDK11 activator that regulates global pre-mRNA splicing.


Asunto(s)
Precursores del ARN , Empalme del ARN , Precursores del ARN/genética , Precursores del ARN/metabolismo , Fosforilación , División Celular , Ciclinas/genética , Ciclinas/metabolismo
7.
Cell ; 149(1): 75-87, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22464324

RESUMEN

Crossovers (COs) between homologous chromosomes ensure their faithful segregation during meiosis. We identify C. elegans COSA-1, a cyclin-related protein conserved in metazoa, as a key component required to convert meiotic double-strand breaks (DSBs) into COs. During late meiotic prophase, COSA-1 localizes to foci that correspond to the single CO site on each homolog pair and indicate sites of eventual concentration of other conserved CO proteins. Chromosomes gain and lose competence to load CO proteins during meiotic progression, with competence to load COSA-1 requiring prior licensing. Our data further suggest a self-reinforcing mechanism maintaining CO designation. Modeling of a nonlinear dose-response relationship between IR-induced DSBs and COSA-1 foci reveals efficient conversion of DSBs into COs when DSBs are limiting and a robust capacity to limit cytologically differentiated CO sites when DSBs are in excess. COSA-1 foci serve as a unique live cell readout for investigating CO formation and CO interference.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/citología , Intercambio Genético , Ciclinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Meiosis , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Cromosomas/metabolismo , Ciclinas/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Modelos Moleculares , Mutación
8.
Mol Cell ; 74(4): 758-770.e4, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30982746

RESUMEN

The cyclin-dependent kinases Cdk4 and Cdk6 form complexes with D-type cyclins to drive cell proliferation. A well-known target of cyclin D-Cdk4,6 is the retinoblastoma protein Rb, which inhibits cell-cycle progression until its inactivation by phosphorylation. However, the role of Rb phosphorylation by cyclin D-Cdk4,6 in cell-cycle progression is unclear because Rb can be phosphorylated by other cyclin-Cdks, and cyclin D-Cdk4,6 has other targets involved in cell division. Here, we show that cyclin D-Cdk4,6 docks one side of an alpha-helix in the Rb C terminus, which is not recognized by cyclins E, A, and B. This helix-based docking mechanism is shared by the p107 and p130 Rb-family members across metazoans. Mutation of the Rb C-terminal helix prevents its phosphorylation, promotes G1 arrest, and enhances Rb's tumor suppressive function. Our work conclusively demonstrates that the cyclin D-Rb interaction drives cell division and expands the diversity of known cyclin-based protein docking mechanisms.


Asunto(s)
Proliferación Celular/genética , Ciclina D/genética , Mapas de Interacción de Proteínas/genética , Proteína de Retinoblastoma/genética , Ciclo Celular/genética , Proteína Sustrato Asociada a CrK/genética , Ciclina D/química , Quinasa 4 Dependiente de la Ciclina/química , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/química , Quinasa 6 Dependiente de la Ciclina/genética , Ciclinas/genética , Fase G1/genética , Humanos , Simulación del Acoplamiento Molecular , Fosforilación/genética , Unión Proteica/genética , Conformación Proteica en Hélice alfa/genética , Proteína de Retinoblastoma/química , Proteína p107 Similar a la del Retinoblastoma/genética , Fase S/genética
9.
Mol Cell ; 74(6): 1264-1277.e7, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31130363

RESUMEN

E2F1, E2F2, and E2F3A, the three activators of the E2F family of transcription factors, are key regulators of the G1/S transition, promoting transcription of hundreds of genes critical for cell-cycle progression. We found that during late S and in G2, the degradation of all three activator E2Fs is controlled by cyclin F, the substrate receptor of 1 of 69 human SCF ubiquitin ligase complexes. E2F1, E2F2, and E2F3A interact with the cyclin box of cyclin F via their conserved N-terminal cyclin binding motifs. In the short term, E2F mutants unable to bind cyclin F remain stable throughout the cell cycle, induce unscheduled transcription in G2 and mitosis, and promote faster entry into the next S phase. However, in the long term, they impair cell fitness. We propose that by restricting E2F activity to the S phase, cyclin F controls one of the main and most critical transcriptional engines of the cell cycle.


Asunto(s)
Ciclo Celular/genética , Ciclinas/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F2/genética , Factor de Transcripción E2F3/genética , Proteínas Ligasas SKP Cullina F-box/genética , Transcripción Genética , Línea Celular Tumoral , Ciclinas/metabolismo , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F2/metabolismo , Factor de Transcripción E2F3/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Aptitud Genética , Células HEK293 , Células HeLa , Humanos , Mutación , Osteoblastos/citología , Osteoblastos/metabolismo , Proteolisis , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal , Ubiquitinación
10.
Mol Cell ; 75(1): 76-89.e3, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31101497

RESUMEN

Cyclin-dependent kinases (CDKs) coordinate hundreds of molecular events during the cell cycle. Multiple cyclins are involved, but the global role of cyclin-specific phosphorylation has remained unsolved. We uncovered a cyclin docking motif, LxF, that mediates binding of replication factor Cdc6 to mitotic cyclin. This interaction leads to phospho-adaptor Cks1-mediated inhibition of M-CDK to facilitate Cdc6 accumulation and sequestration in mitosis. The LxF motif and Cks1 also mediate the mutual inhibition between M-CDK and the tyrosine kinase Swe1. Additionally, the LxF motif is critical for targeting M-CDK to phosphorylate several mitotic regulators; for example, Spo12 is targeted via LxF to release the phosphatase Cdc14. The results complete the full set of G1, S, and M-CDK docking mechanisms and outline the unified role of cyclin specificity and CDK activity thresholds. Cooperation of cyclin and Cks1 docking creates a variety of CDK thresholds and switching orders, including combinations of last in, first out (LIFO) and first in, first out (FIFO) ordering.


Asunto(s)
Proteínas de Ciclo Celular/genética , Ciclinas/genética , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Regulación Fúngica de la Expresión Génica , Puntos de Control de la Fase M del Ciclo Celular/genética , Puntos de Control de la Fase S del Ciclo Celular/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Sitios de Unión , Proteínas de Ciclo Celular/metabolismo , Ciclinas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal
11.
Genes Dev ; 33(7-8): 418-435, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30819820

RESUMEN

The RNA polymerase II (RNAPII) C-terminal domain kinase, CDK12, regulates genome stability, expression of DNA repair genes, and cancer cell resistance to chemotherapy and immunotherapy. In addition to its role in mRNA biosynthesis of DNA repair genes, we show here that CDK12 phosphorylates the mRNA 5' cap-binding repressor, 4E-BP1, to promote translation of mTORC1-dependent mRNAs. In particular, we found that phosphorylation of 4E-BP1 by mTORC1 (T37 and T46) facilitates subsequent CDK12 phosphorylation at two Ser-Pro sites (S65 and T70) that control the exchange of 4E-BP1 with eIF4G at the 5' cap of CHK1 and other target mRNAs. RNA immunoprecipitation coupled with deep sequencing (RIP-seq) revealed that CDK12 regulates release of 4E-BP1, and binding of eIF4G, to many mTORC1 target mRNAs, including those needed for MYC transformation. Genome-wide ribosome profiling (Ribo-seq) further identified specific CDK12 "translation-only" target mRNAs, including many mTORC1 target mRNAs as well as many subunits of mitotic and centromere/centrosome complexes. Accordingly, confocal imaging analyses revealed severe chromosome misalignment, bridging, and segregation defects in cells deprived of CDK12 or CCNK. We conclude that the nuclear RNAPII-CTD kinase CDK12 cooperates with mTORC1, and controls a specialized translation network that is essential for mitotic chromosome stability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasas Ciclina-Dependientes/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Inestabilidad Genómica/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Ciclinas/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Humanos , Mitosis/genética , Fosforilación/genética , Unión Proteica/genética
12.
Development ; 150(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38014633

RESUMEN

Sall1 and Sall4 (Sall1/4), zinc-finger transcription factors, are expressed in the progenitors of the second heart field (SHF) and in cardiomyocytes during the early stages of mouse development. To understand the function of Sall1/4 in heart development, we generated heart-specific Sall1/4 functionally inhibited mice by forced expression of the truncated form of Sall4 (ΔSall4) in the heart. The ΔSall4-overexpression mice exhibited a hypoplastic right ventricle and outflow tract, both of which were derived from the SHF, and a thinner ventricular wall. We found that the numbers of proliferative SHF progenitors and cardiomyocytes were reduced in ΔSall4-overexpression mice. RNA-sequencing data showed that Sall1/4 act upstream of the cyclin-dependent kinase (CDK) and cyclin genes, and of key transcription factor genes for the development of compact cardiomyocytes, including myocardin (Myocd) and serum response factor (Srf). In addition, ChIP-sequencing and co-immunoprecipitation analyses revealed that Sall4 and Myocd form a transcriptional complex with SRF, and directly bind to the upstream regulatory regions of the CDK and cyclin genes (Cdk1 and Ccnb1). These results suggest that Sall1/4 are critical for the proliferation of cardiac cells via regulation of CDK and cyclin genes that interact with Myocd and SRF.


Asunto(s)
Quinasas Ciclina-Dependientes , Miocitos Cardíacos , Animales , Ratones , Proliferación Celular/genética , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Miocitos Cardíacos/metabolismo , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción/metabolismo
13.
EMBO Rep ; 25(2): 745-769, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38233717

RESUMEN

Pho85 is a multifunctional CDK that signals to the cell when environmental conditions are favorable. It has been connected to cell cycle control, mainly in Start where it promotes the G1/S transition. Here we describe that the Start repressor Whi7 is a key target of Pho85 in the regulation of cell cycle entry. The phosphorylation of Whi7 by Pho85 inhibits the repressor and explains most of the contribution of the CDK in the activation of Start. Mechanistically, Pho85 downregulates Whi7 protein levels through the control of Whi7 protein stability and WHI7 gene transcription. Whi7 phosphorylation by Pho85 also restrains the intrinsic ability of Whi7 to associate with promoters. Furthermore, although Whi5 is the main Start repressor in normal cycling cells, in the absence of Pho85, Whi7 becomes the major repressor leading to G1 arrest. Overall, our results reveal a novel mechanism by which Pho85 promotes Start through the regulation of the Whi7 repressor at multiple levels, which may confer to Whi7 a functional specialization to connect the response to adverse conditions with the cell cycle control.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomycetales , Ciclo Celular/genética , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Regulación Fúngica de la Expresión Génica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomycetales/metabolismo
14.
Mol Cell ; 69(6): 938-952.e6, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29547722

RESUMEN

We report an unanticipated system of joint regulation by cyclin-dependent kinase (CDK) and mitogen-activated protein kinase (MAPK), involving collaborative multi-site phosphorylation of a single substrate. In budding yeast, the protein Ste5 controls signaling through a G1 arrest pathway. Upon cell-cycle entry, CDK inhibits Ste5 via multiple phosphorylation sites, disrupting its membrane association. Using quantitative time-lapse microscopy, we examined Ste5 membrane recruitment dynamics at different cell-cycle stages. Surprisingly, in S phase, where Ste5 recruitment should be blocked, we observed an initial recruitment followed by a steep drop-off. This delayed inhibition revealed a requirement for both CDK activity and negative feedback from the pathway MAPK Fus3. Mutagenesis, mass spectrometry, and electrophoretic analyses suggest that the CDK and MAPK modify shared sites, which are most extensively phosphorylated when both kinases are active and able to bind their docking sites on Ste5. Such collaborative phosphorylation can broaden regulatory inputs and diversify output dynamics of signaling pathways.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Sitios de Unión , Puntos de Control del Ciclo Celular , Membrana Celular/enzimología , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Ciclinas/metabolismo , Cinética , Proteínas Quinasas Activadas por Mitógenos/genética , Fosforilación , Unión Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Especificidad por Sustrato
15.
Mol Cell ; 70(4): 628-638.e5, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29775579

RESUMEN

Cell survival to replication stress depends on the activation of the Mec1ATR-Rad53 checkpoint response that protects the integrity of stalled forks and controls the origin firing program. Here we found that Mad2, a member of the spindle assembly checkpoint (SAC), contributes to efficient origin firing and to cell survival in response to replication stress. We show that Rad53 and Mad2 promote S-phase cyclin expression through different mechanisms: while Rad53 influences Clb5,6 degradation, Mad2 promotes their protein synthesis. We found that Mad2 co-sediments with polysomes and modulates the association of the translation inhibitor Caf204E-BP with the translation machinery and the initiation factor eIF4E. This Mad2-dependent translational regulatory process does not depend on other SAC proteins. Altogether our observations indicate that Mad2 has an additional function outside of mitosis to control DNA synthesis and collaborates with the Mec1-Rad53 regulatory axis to allow cell survival in response to replication stress.


Asunto(s)
Ciclinas/genética , Replicación del ADN , Proteínas Mad2/metabolismo , Mitosis , Biosíntesis de Proteínas , Fase S , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , Ciclina B/genética , Ciclina B/metabolismo , Ciclinas/metabolismo , Proteínas Mad2/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Origen de Réplica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Nucleic Acids Res ; 52(3): 1258-1271, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38048302

RESUMEN

Progression through the mitotic and meiotic cell cycle is driven by fluctuations in the levels of cyclins, the regulatory subunits controlling the localization and activity of CDK1 kinases. Cyclin levels are regulated through a precise balance of synthesis and degradation. Here we demonstrate that the synthesis of Cyclin B1 during the oocyte meiotic cell cycle is defined by the selective translation of mRNA variants generated through alternative cleavage and polyadenylation (APA). Using gene editing in mice, we introduced mutations into the proximal and distal polyadenylation elements of the 3' untranslated region (UTR) of the Ccnb1 mRNA. Through in vivo loss-of-function experiments, we demonstrate that the translation of mRNA with a short 3' UTR specifies Cyclin B1 protein levels that set the timing of meiotic re-entry. In contrast, translation directed by a long 3' UTR is necessary to direct Cyclin B1 protein accumulation during the MI/MII transition. These findings establish that the progression through the cell cycle is dependent on the selective translation of multiple mRNA variants generated by APA.


Asunto(s)
Ciclina B1 , Meiosis , Poliadenilación , Animales , Ratones , Regiones no Traducidas 3'/genética , Ciclo Celular/genética , Ciclina B1/genética , Ciclina B1/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Oocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
PLoS Genet ; 19(1): e1010602, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36652497

RESUMEN

Silkworm silk gland cells undergo endoreplicating cycle and rapid growth during the larval period, and synthesize massive silk proteins for silk production. In this study, we demonstrated that a binary transgenic CRISPR/Cas9 approach-mediated Fzr mutation in silkworm posterior silk gland (PSG) cells caused an arrest of silk gland growth and a decrease in silk production. Mechanistically, PSG-specific Fzr mutation blocked endoreplication progression by inducing an expression dysregulation of several cyclin proteins and DNA replication-related regulators. Moreover, based on label-free quantitative proteome analysis, we showed in PSG cells that Fzr mutation-induced decrease in the levels of cyclin proteins and silk proteins was likely due to an inhibition of the ribosome biogenesis pathway associated with mRNA translation, and/or an enhance of the ubiquitin-mediated protein degradation pathway. Rbin-1 inhibitor-mediated blocking of ribosomal biogenesis pathway decreased DNA replication in PSG cells and silk production. Altogether, our results reveal that Fzr positively regulates PSG growth and silk production in silkworm by promoting endoreplication and protein synthesis in PSG cells.


Asunto(s)
Bombyx , Animales , Endorreduplicación , Seda/genética , Biosíntesis de Proteínas/genética , Ciclinas/genética , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo
18.
Semin Cancer Biol ; 98: 51-63, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38135020

RESUMEN

CDK4, along with its regulatory subunit, cyclin D, drives the transition from G1 to S phase, during which DNA replication and metabolic activation occur. In this canonical pathway, CDK4 is essentially a transcriptional regulator that acts through phosphorylation of retinoblastoma protein (RB) and subsequent activation of the transcription factor E2F, ultimately triggering the expression of genes involved in DNA synthesis and cell cycle progression to S phase. In this review, we focus on the newly reported functions of CDK4, which go beyond direct regulation of the cell cycle. In particular, we describe the extranuclear roles of CDK4, including its roles in the regulation of metabolism, cell fate, cell dynamics and the tumor microenvironment. We describe direct phosphorylation targets of CDK4 and decipher how CDK4 influences these physiological processes in the context of cancer.


Asunto(s)
Ciclinas , Neoplasias , Humanos , Ciclinas/genética , Ciclinas/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Ciclina D/metabolismo , Fosforilación , Ciclo Celular/genética , Neoplasias/genética , Proteínas de Ciclo Celular/metabolismo , Microambiente Tumoral
19.
Hum Mol Genet ; 32(14): 2386-2398, 2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37220877

RESUMEN

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are fatal neurodegenerative disorders that share pathological features, including the aberrant accumulation of ubiquitinated protein inclusions within motor neurons. Previously, we have shown that the sequestration of ubiquitin (Ub) into inclusions disrupts Ub homeostasis in cells expressing ALS-associated variants superoxide dismutase 1 (SOD1), fused in sarcoma (FUS) and TAR DNA-binding protein 43 (TDP-43). Here, we investigated whether an ALS/FTD-linked pathogenic variant in the CCNF gene, encoding the E3 Ub ligase Cyclin F (CCNF), also perturbs Ub homeostasis. The presence of a pathogenic CCNF variant was shown to cause ubiquitin-proteasome system (UPS) dysfunction in induced pluripotent stem cell-derived motor neurons harboring the CCNF  S621G mutation. The expression of the CCNFS621G variant was associated with an increased abundance of ubiquitinated proteins and significant changes in the ubiquitination of key UPS components. To further investigate the mechanisms responsible for this UPS dysfunction, we overexpressed CCNF in NSC-34 cells and found that the overexpression of both wild-type (WT) and the pathogenic variant of CCNF (CCNFS621G) altered free Ub levels. Furthermore, double mutants designed to decrease the ability of CCNF to form an active E3 Ub ligase complex significantly improved UPS function in cells expressing both CCNFWT and the CCNFS621G variant and were associated with increased levels of free monomeric Ub. Collectively, these results suggest that alterations to the ligase activity of the CCNF complex and the subsequent disruption to Ub homeostasis play an important role in the pathogenesis of CCNF-associated ALS/FTD.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedad de Pick , Humanos , Esclerosis Amiotrófica Lateral/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Ciclinas/genética , Neuronas Motoras/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Enfermedad de Pick/metabolismo , Homeostasis/genética , Mutación
20.
EMBO J ; 40(19): e108041, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34431536

RESUMEN

The role of WNT/ß-catenin signalling in mouse neocortex development remains ambiguous. Most studies demonstrate that WNT/ß-catenin regulates progenitor self-renewal but others suggest it can also promote differentiation. Here we explore the role of WNT/STOP signalling, which stabilizes proteins during G2/M by inhibiting glycogen synthase kinase (GSK3)-mediated protein degradation. We show that mice mutant for cyclin Y and cyclin Y-like 1 (Ccny/l1), key regulators of WNT/STOP signalling, display reduced neurogenesis in the developing neocortex. Specifically, basal progenitors, which exhibit delayed cell cycle progression, were drastically decreased. Ccny/l1-deficient apical progenitors show reduced asymmetric division due to an increase in apical-basal astral microtubules. We identify the neurogenic transcription factors Sox4 and Sox11 as direct GSK3 targets that are stabilized by WNT/STOP signalling in basal progenitors during mitosis and that promote neuron generation. Our work reveals that WNT/STOP signalling drives cortical neurogenesis and identifies mitosis as a critical phase for neural progenitor fate.


Asunto(s)
Mitosis , Neocórtex/embriología , Neocórtex/metabolismo , Neurogénesis , Vía de Señalización Wnt , Secuencia de Aminoácidos , Animales , Biomarcadores , Ciclo Celular , Diferenciación Celular/genética , Ciclinas/genética , Ciclinas/metabolismo , Embrión de Mamíferos , Técnica del Anticuerpo Fluorescente , Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Mitosis/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Fosforilación , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA