Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 14677, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37674027

RESUMO

Reactive oxygen species (ROS) are an important source of cellular damage. When ROS intracellular levels increase, oxidative stress takes place affecting DNA stability and metabolic functions. To prevent these effects, stress-activated protein kinases (SAPKs) delay cell cycle progression and induce a transcriptional response that activates antioxidant mechanisms ensuring cell adaptation and survival. Fission yeast Cdc14-like phosphatase Flp1 (also known as Clp1) has a well-established role in cell cycle regulation. Moreover, Flp1 contributes to checkpoint activation during replication stress. Here, we show that Flp1 has a role in fine-tuning the cellular oxidative stress response. Phosphorylation-dependent nucleolar release of Flp1 in response to oxidative stress conditions plays a role in the cellular transcriptional response. Thus, Flp1 ablation increases the transcriptional response to oxidative stress, in both intensity and duration, upregulating both Atf1/Pcr1- and Pap1-dependent stress induced genes. Remarkably, we found that Flp1 interacts with the Atf1/Pcr1 complex with Pcr1 acting as a direct substrate. Our results provide evidence that Flp1 modulates the oxidative stress response by limiting the Atf1/Pcr1-mediated transcription.


Assuntos
Schizosaccharomyces , Schizosaccharomyces/genética , Espécies Reativas de Oxigênio , Estresse Oxidativo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Monoéster Fosfórico Hidrolases
2.
Genes (Basel) ; 11(2)2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32093406

RESUMO

Fidelity in chromosome duplication and segregation is indispensable for maintaining genomic stability and the perpetuation of life. Challenges to genome integrity jeopardize cell survival and are at the root of different types of pathologies, such as cancer. The following three main sources of genomic instability exist: DNA damage, replicative stress, and chromosome segregation defects. In response to these challenges, eukaryotic cells have evolved control mechanisms, also known as checkpoint systems, which sense under-replicated or damaged DNA and activate specialized DNA repair machineries. Cells make use of these checkpoints throughout interphase to shield genome integrity before mitosis. Later on, when the cells enter into mitosis, the spindle assembly checkpoint (SAC) is activated and remains active until the chromosomes are properly attached to the spindle apparatus to ensure an equal segregation among daughter cells. All of these processes are tightly interconnected and under strict regulation in the context of the cell division cycle. The chromosomal instability underlying cancer pathogenesis has recently emerged as a major source for understanding the mitotic processes that helps to safeguard genome integrity. Here, we review the special interconnection between the S-phase and mitosis in the presence of under-replicated DNA regions. Furthermore, we discuss what is known about the DNA damage response activated in mitosis that preserves chromosomal integrity.


Assuntos
Instabilidade Genômica/genética , Instabilidade Genômica/fisiologia , Proteínas de Ciclo Celular/genética , Instabilidade Cromossômica/genética , Segregação de Cromossomos/genética , Cromossomos/genética , Dano ao DNA/genética , Reparo do DNA/genética , Replicação do DNA/genética , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Mitose/genética , Fase S/genética , Fuso Acromático/genética
3.
Cell Rep ; 29(5): 1323-1335.e5, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31665643

RESUMO

DNA damage tolerance plays a key role in protecting cell viability through translesion synthesis and template switching-mediated bypass of genotoxic polymerase-blocking base lesions. Both tolerance pathways critically rely on ubiquitylation of the proliferating-cell nuclear antigen (PCNA) on lysine 164 and have been proposed to operate uncoupled from replication. We report that Ubp10 and Ubp12 ubiquitin proteases differentially cooperate in PCNA deubiquitylation, owing to distinct activities on PCNA-linked ubiquitin chains. Ubp10 and Ubp12 associate with replication forks in a fashion determined by Ubp10 dependency on lagging-strand PCNA residence, and they downregulate translesion polymerase recruitment and template switch events engaging nascent strands. These findings reveal PCNAK164 deubiquitylation as a key mechanism for the modulation of lesion bypass during replication, which might set a framework for establishing strand-differential pathway choices. We propose that damage tolerance is tempered at replication forks to limit the extension of bypass events and sustain chromosome replication rates.


Assuntos
Dano ao DNA , Replicação do DNA , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação , DNA Fúngico/biossíntese , DNA Polimerase Dirigida por DNA/metabolismo , Mutação/genética , Fase S , Moldes Genéticos
4.
PLoS Genet ; 15(2): e1007981, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30807579

RESUMO

Gene expression is generally regulated by recruitment of transcription factors and RNA polymerase II (RNAP II) to specific sequences in the gene promoter region. The Integrator complex mediates processing of small nuclear RNAs (snRNAs) as well as the initiation and release of paused RNAP II at specific genes in response to growth factors. Here we show that in C. elegans, disruption of the Integrator complex leads to transcription of genes located downstream of the snRNA loci via a non-conventional transcription mechanism based on the lack of processing of the snRNAs. RNAP II read-through generates long chimeric RNAs containing snRNA, the intergenic region and the mature mRNA of the downstream gene located in sense. These chimeric sn-mRNAs remain as untranslated long non-coding RNAs, in the case of U1- and U2-derived sn-mRNAs, but can be translated to proteins in the case of SL-derived sn-mRNAs. The transcriptional effect caused by disruption of the Integrator complex is not restricted to genes located downstream of the snRNA loci but also affects key regulators of signal transduction such as kinases and phosphatases. Our findings highlight that these transcriptional alterations may be behind the correlation between mutations in the Integrator complex and tumor transformation.


Assuntos
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , RNA de Helmintos/genética , RNA de Helmintos/metabolismo , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/embriologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Regulação para Baixo , Técnicas de Silenciamento de Genes , Genes de Helmintos , Mutação , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Processamento Pós-Transcricional do RNA , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Regulação para Cima
5.
J Cell Biol ; 218(2): 541-558, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30626720

RESUMO

Greatwall (GWL) is an essential kinase that indirectly controls PP2A-B55, the phosphatase counterbalancing cyclin B/CDK1 activity during mitosis. In Xenopus laevis egg extracts, GWL-mediated phosphorylation of overexpressed ARPP19 and ENSA turns them into potent PP2A-B55 inhibitors. It has been shown that the GWL/ENSA/PP2A-B55 axis contributes to the control of DNA replication, but little is known about the role of ARPP19 in cell division. By using conditional knockout mouse models, we investigated the specific roles of ARPP19 and ENSA in cell division. We found that Arpp19, but not Ensa, is essential for mouse embryogenesis. Moreover, Arpp19 ablation dramatically decreased mouse embryonic fibroblast (MEF) viability by perturbing the temporal pattern of protein dephosphorylation during mitotic progression, possibly by a drop of PP2A-B55 activity inhibition. We show that these alterations are not prevented by ENSA, which is still expressed in Arpp19 Δ/Δ MEFs, suggesting that ARPP19 is essential for mitotic division. Strikingly, we demonstrate that unlike ARPP19, ENSA is not required for early embryonic development. Arpp19 knockout did not perturb the S phase, unlike Ensa gene ablation. We conclude that, during mouse embryogenesis, the Arpp19 and Ensa paralog genes display specific functions by differentially controlling cell cycle progression.


Assuntos
Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mitose/fisiologia , Fosfoproteínas/metabolismo , Fase S/fisiologia , Animais , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/fisiologia , Fibroblastos/citologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Knockout , Fosfoproteínas/genética , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Xenopus laevis
6.
Sci Rep ; 8(1): 11871, 2018 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30089874

RESUMO

Cdc14 enzymes compose a family of highly conserved phosphatases that are present in a wide range of organisms, including yeast and humans, and that preferentially reverse the phosphorylation of Cyclin-Dependent Kinase (Cdk) substrates. The budding yeast Cdc14 orthologue has essential functions in the control of late mitosis and cytokinesis. In mammals, however, the two Cdc14 homologues, Cdc14A and Cdc14B, do not play a prominent role in controlling late mitotic events, suggesting that some Cdc14 functions are not conserved across species. Moreover, in yeast, Cdc14 is regulated by changes in its subcellular location and by phosphorylation events. In contrast, little is known about the regulation of human Cdc14 phosphatases. Here, we have studied how the human Cdc14A orthologue is regulated during the cell cycle. We found that Cdc14A is phosphorylated on Ser411, Ser453 and Ser549 by Cdk1 early in mitosis and becomes dephosphorylated during late mitotic stages. Interestingly, in vivo and in vitro experiments revealed that, unlike in yeast, Cdk1-mediated phosphorylation of human Cdc14A did not control its catalytic activity but likely modulated its interaction with other proteins in early mitosis. These findings point to differences in Cdk1-mediated mechanisms of regulation between human and yeast Cdc14 orthologues.


Assuntos
Aminoácidos/metabolismo , Proteína Quinase CDC2/metabolismo , Ciclo Celular/fisiologia , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação/fisiologia , Fenômenos Bioquímicos/fisiologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Citocinese/fisiologia , Proteínas Fúngicas/metabolismo , Células HEK293 , Células HeLa , Humanos , Mitose/fisiologia , Proteínas Tirosina Fosfatases , Leveduras/metabolismo
7.
Nat Commun ; 7: 11821, 2016 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27273576

RESUMO

A coding polymorphism of human ATG16L1 (rs2241880; T300A) increases the risk of Crohn's disease and it has been shown to enhance susceptibility of ATG16L1 to caspase cleavage. Here we show that T300A also alters the ability of the C-terminal WD40-repeat domain of ATG16L1 to interact with an amino acid motif that recognizes this region. Such alteration impairs the unconventional autophagic activity of TMEM59, a transmembrane protein that contains the WD40 domain-binding motif, and disrupts its normal intracellular trafficking and its ability to engage ATG16L1 in response to bacterial infection. TMEM59-induced autophagy is blunted in cells expressing the fragments generated by caspase processing of the ATG16L1-T300A risk allele, whereas canonical autophagy remains unaffected. These results suggest that the T300A polymorphism alters the function of motif-containing molecules that engage ATG16L1 through the WD40 domain, either by influencing this interaction under non-stressful conditions or by inhibiting their downstream autophagic signalling after caspase-mediated cleavage.


Assuntos
Proteínas Relacionadas à Autofagia/química , Proteínas Relacionadas à Autofagia/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Doença de Crohn/genética , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único/genética , Repetições WD40 , Alelos , Motivos de Aminoácidos , Animais , Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Proteínas de Transporte/metabolismo , Caspase 3/metabolismo , Células HCT116 , Células HEK293 , Humanos , Espaço Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Ligação Proteica , Transporte Proteico , Fatores de Risco , Staphylococcus aureus/fisiologia
8.
Sci Rep ; 6: 25513, 2016 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-27151298

RESUMO

Proliferating-cell nuclear antigen (PCNA) is a DNA sliding clamp with an essential function in DNA replication and a key role in tolerance to DNA damage by ensuring the bypass of lesions. In eukaryotes, DNA damage tolerance is regulated by ubiquitylation of lysine 164 of PCNA through a well-known control mechanism; however, the regulation of PCNA deubiquitylation remains poorly understood. Our work is a systematic and functional study on PCNA deubiquitylating enzymes (DUBs) in Schizosaccharomyces pombe. Our study reveals that the deubiquitylation of PCNA in fission yeast cells is a complex process that requires several ubiquitin proteases dedicated to the deubiquitylation of a specific subnuclear fraction of mono- and di-ubiquitylated PCNA or a particular type of poly-ubiquitylated PCNA and that there is little redundancy among these enzymes. To understand how DUB activity regulates the oscillatory pattern of ubiquitylated PCNA in fission yeast, we assembled multiple DUB mutants and found that a quadruple mutation of ubp2(+), ubp12(+), ubp15(+), and ubp16(+) leads to the stable accumulation of mono-, di-, and poly-ubiquitylated forms of PCNA, increases S-phase duration, and sensitizes cells to DNA damage. Our data suggest that the dynamic ubiquitylation and deubiquitylation of PCNA occurs during S-phase to ensure processive DNA replication.


Assuntos
Ciclo Celular , Enzimas Desubiquitinantes/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Processamento de Proteína Pós-Traducional , Schizosaccharomyces/fisiologia , Replicação do DNA , Enzimas Desubiquitinantes/genética , Técnicas de Inativação de Genes , Schizosaccharomyces/metabolismo
9.
Mol Biol Cell ; 23(23): 4515-25, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23051732

RESUMO

The activity of Cdk1-cyclin B1 mitotic complexes is regulated by the balance between the counteracting activities of Wee1/Myt1 kinases and Cdc25 phosphatases. These kinases and phosphatases must be strictly regulated to ensure proper mitotic timing. One masterpiece of this regulatory network is Cdk1, which promotes Cdc25 activity and suppresses inhibitory Wee1/Myt1 kinases through direct phosphorylation. The Cdk1-dependent phosphorylation of Wee1 primes phosphorylation by additional kinases such as Plk1, triggering Wee1 degradation at the onset of mitosis. Here we report that Cdc14A plays an important role in the regulation of Wee1 stability. Depletion of Cdc14A results in a significant reduction in Wee1 protein levels. Cdc14A binds to Wee1 at its amino-terminal domain and reverses CDK-mediated Wee1 phosphorylation. In particular, we found that Cdc14A inhibits Wee1 degradation through the dephosphorylation of Ser-123 and Ser-139 residues. Thus the lack of phosphorylation of these two residues prevents the interaction with Plk1 and the consequent efficient Wee1 degradation at the onset of mitosis. These data support the hypothesis that Cdc14A counteracts Cdk1-cyclin B1 activity through Wee1 dephosphorylation.


Assuntos
Proteínas de Ciclo Celular , Mitose/genética , Proteínas Nucleares , Monoéster Fosfórico Hidrolases , Fosforilação , Proteínas Tirosina Quinases , Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ciclina B1/metabolismo , Regulação da Expressão Gênica , Células HCT116 , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Estabilidade Proteica , Proteínas Tirosina Fosfatases , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteólise , Proteínas Proto-Oncogênicas/metabolismo , Fosfatases cdc25/metabolismo , Quinase 1 Polo-Like
10.
Nat Cell Biol ; 13(12): 1450-6, 2011 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-22020438

RESUMO

Kinases and phosphatases regulate messenger RNA synthesis through post-translational modification of the carboxy-terminal domain (CTD) of the largest subunit of RNA polymerase II (ref. 1). In yeast, the phosphatase Cdc14 is required for mitotic exit(2,3) and for segregation of repetitive regions(4). Cdc14 is also a subunit of the silencing complex RENT (refs 5,6), but no roles in transcriptional repression have been described. Here we report that inactivation of Cdc14 causes silencing defects at the intergenic spacer sequences of ribosomal genes during interphase and at Y' repeats in subtelomeric regions during mitosis. We show that the role of Cdc14 in silencing is independent of the RENT deacetylase subunit Sir2. Instead, Cdc14 acts directly on RNA polymerase II by targeting CTD phosphorylation at Ser 2 and Ser 5. We also find that the role of Cdc14 as a CTD phosphatase is conserved in humans. Finally, telomere segregation defects in cdc14 mutants(4) correlate with the presence of subtelomeric Y' elements and can be rescued by transcriptional inhibition of RNA polymerase II.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Inativação Gênica/fisiologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , RNA Polimerase II/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Telômero/metabolismo , Transcrição Gênica/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , DNA Espaçador Ribossômico/genética , Interfase/genética , Mitose/genética , Fosforilação/genética , Proteínas Tirosina Fosfatases/antagonistas & inibidores , RNA Polimerase II/antagonistas & inibidores , Sequências Repetitivas de Ácido Nucleico , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/enzimologia , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Supressão Genética/fisiologia , Telômero/enzimologia
11.
Cell Cycle ; 10(3): 387-91, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21233601

RESUMO

Cdc14 belongs to a dual-specificity phosphatase family highly conserved through evolution that preferentially reverses CDK (Cyclin dependent kinases) -dependent phosphorylation events. In the yeast Saccharomyces cerevisiae, Cdc14 is an essential regulator of late mitotic events and exit from mitosis by counteracting CDK activity at the end of mitosis. However, many studies have shown that Cdc14 is dispensable for exiting mitosis in all other model systems analyzed. In fission yeast, the Cdc14 homologue Flp1/Clp1 regulates the stability of the mitotic inducer Cdc25 at the end of mitosis to ensure Cdk1 inactivation before cytokinesis. We have recently reported that human Cdc14A, the Cdc14 isoform located at the centrosomes during interphase, down-regulates Cdc25 activity at the G2/M transition to prevent premature activation of Cdk1-Cyclin B1 complexes and untimely entry into mitosis. Here we speculate about new molecular mechanisms for Cdc14A and discuss the current evidence suggesting that Cdc14 phosphatase plays a role in cell cycle control in higher eukaryotes.


Assuntos
Proteína Quinase CDC2/antagonistas & inibidores , Divisão Celular , Fase G2 , Genes cdc , Monoéster Fosfórico Hidrolases/fisiologia , Centrossomo/metabolismo , Humanos , Modelos Genéticos , Monoéster Fosfórico Hidrolases/genética , Fosforilação , Proteínas Tirosina Fosfatases
12.
Sci Rep ; 1: 189, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22355704

RESUMO

Cdc14 is an essential phosphatase in yeast but its role in the mammalian cell cycle remains obscure. We report here that Cdc14b-knockout cells display unscheduled induction of multiple cell cycle regulators resulting in early entry into DNA replication and mitosis from quiescence. Cdc14b dephosphorylates Ser5 at the C-terminal domain (CTD) of RNA polymerase II, a major substrate of cyclin-dependent kinases. Lack of Cdc14b results in increased CTD-Ser5 phosphorylation, epigenetic modifications that mark active chromatin, and transcriptional induction of cell cycle regulators. These data suggest a function for mammalian Cdc14 phosphatases in the control of transcription during the cell cycle.


Assuntos
Ciclo Celular , Fosfatases de Especificidade Dupla/metabolismo , RNA Polimerase II/metabolismo , Transcrição Gênica , Alelos , Animais , Sítios de Ligação , Células Cultivadas , Epigênese Genética , Éxons , Fibroblastos/citologia , Regulação Enzimológica da Expressão Gênica , Camundongos , Mitose , Fosfatos/química , Fosforilação , Ativação Transcricional
13.
J Biol Chem ; 285(52): 40544-53, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20956543

RESUMO

The Cdc14 family of serine-threonine phosphatases antagonizes CDK activity by reversing CDK-dependent phosphorylation events. It is well established that the yeast members of this family bring about the M/G1 transition. Budding yeast Cdc14 is essential for CDK inactivation at the end of mitosis and fission yeast Cdc14 homologue Flp1/Clp1 down-regulates Cdc25 to ensure the inactivation of mitotic CDK complexes to trigger cell division. However, the functions of human Cdc14 homologues remain poorly understood. Here we have tested the hypothesis that Cdc14A might regulate Cdc25 mitotic inducers in human cells. We found that increasing levels of Cdc14A delay entry into mitosis by inhibiting Cdk1-cyclin B1 activity. By contrast, lowering the levels of Cdc14A accelerates mitotic entry. Biochemical analyses revealed that Cdc14A acts through key Cdk1-cyclin B1 regulators. We observed that Cdc14A directly bound to and dephosphorylated Cdc25B, inhibiting its catalytic activity. Cdc14A also regulated the activity of Cdc25A at the G2/M transition. Our results indicate that Cdc14A phosphatase prevents premature activation of Cdk1 regulating Cdc25A and Cdc25B at the entry into mitosis.


Assuntos
Fase G2/fisiologia , Mitose/fisiologia , Monoéster Fosfórico Hidrolases/metabolismo , Fosfatases cdc25/metabolismo , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Linhagem Celular , Ativação Enzimática/fisiologia , Humanos , Monoéster Fosfórico Hidrolases/genética , Proteínas Tirosina Fosfatases , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Fosfatases cdc25/genética
14.
Cell Cycle ; 5(24): 2894-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17172867

RESUMO

Human Cdc14A is an evolutionary conserved dual-specificity protein phosphatase that reverses the modifications effected by cyclin-dependent kinases and plays an important role in centrosome duplication and mitotic regulation. Few substrates of Cdc14A have been identified, some of them with homologues in yeast that, in turn, are substrates of the Saccharomyces cerevisiae Cdc14 homologue, a protein phosphatase essential for yeast cell viability owing its role in mitotic exit regulation. Identification of the physiological substrates of human Cdc14A is an immediate goal in order to elucidate which cellular processes it regulates. Here, we show that human Cdc14A can dephosphorylate Cdc25A in vitro. Specifically, the Cdk1/Cyclin-B1-dependent phosphate groups on Ser115 and Ser320 of Cdc25A were found to be removed by Cdc14A. Cdc25A is an important cell cycle-regulatory protein involved in several cell cycle transitions and checkpoint responses and whose function and own regulation depend on complex phosphorylation/dephosphorylation-mediated processes. Importantly, we also show that the upregulation of Cdc14A phosphatase affects Cdc25A protein levels in human cells. Our results suggest that Cdc14A may be involved in the cell cycle regulation of Cdc25A stability.


Assuntos
Proteína Quinase CDC2/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosfosserina/metabolismo , Fosfatases cdc25/química , Fosfatases cdc25/metabolismo , Sequência de Aminoácidos , Ciclina B/metabolismo , Ciclina B1 , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Fosforilação , Proteínas Tirosina Fosfatases
15.
J Biol Chem ; 280(32): 29144-50, 2005 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15911625

RESUMO

Budding and fission yeast Cdc14 homologues, a conserved family of serine-threonine phosphatases, play a role in the inactivation of mitotic cyclin-dependent kinases (CDKs) by molecularly distinct mechanisms. Saccharomyces cerevisiae Cdc14 protein phosphatase inactivates CDKs by promoting mitotic cyclin degradation and the accumulation of a CDK inhibitor to allow budding yeast cells to exit from mitosis. Schizosaccharomyces pombe Flp1 phosphatase down-regulates CDK/cyclin activity, controlling the degradation of the Cdc25 tyrosine phosphatase for fission yeast cells to undergo cytokinesis. In the present work, we show that human Cdc14 homologues (hCdc14A and hCdc14B) rescued flp1-deficient fission yeast strains, indicating functional homology. We also show that hCdc14A and B interacted in vivo with S. pombe Cdc25 and that hCdc14A dephosphorylated this mitotic inducer both in vitro and in vivo. Our results support a Cdc14 conserved inhibitory mechanism acting on S. pombe Cdc25 protein and suggest that human cells may regulate Cdc25 in a similar manner to inactivate Cdk1-mitotic cyclin complexes.


Assuntos
Fosfoproteínas Fosfatases/química , Monoéster Fosfórico Hidrolases/química , Proteínas de Saccharomyces cerevisiae/química , Schizosaccharomyces/enzimologia , Western Blotting , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Citocinese , DNA/química , Regulação para Baixo , Citometria de Fluxo , Teste de Complementação Genética , Glutationa Transferase/metabolismo , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Mitose , Fenótipo , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Fuso Acromático , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...