Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2502: 271-282, 2022.
Article in English | MEDLINE | ID: mdl-35412245

ABSTRACT

We describe a method for rapid identification of protein kinase substrates within the nuclear envelope. Open mitosis in higher eukaryotes is characterized by nuclear envelope breakdown (NEBD) concerted with disassembly of the nuclear lamina and dissociation of nuclear pore complexes (NPCs) into individual subcomplexes. Evidence indicates that reversible phosphorylation events largely drive this mitotic NEBD. These posttranslational modifications likely disrupt structurally significant interactions among nucleoporins (Nups), lamina and membrane proteins of the nuclear envelope (NE). It is therefore critical to determine when and where these substrates are phosphorylated. One likely regulator is the mitotic kinase: Cdk1-Cyclin B. We employed an "analog-sensitive" Cdk1 to bio-orthogonally and uniquely label its substrates in the NE with a phosphate analog tag. Subsequently, peptides covalently modified with the phosphate analogs are rapidly purified by a tag-specific covalent capture and release methodology. In this manner, we were able to confirm the identity of known Cdk1 targets in the NE and discover additional candidates for regulation by mitotic phosphorylation.


Subject(s)
Nuclear Envelope , Phosphopeptides , Animals , Cyclin B/chemistry , Cyclin B/metabolism , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , HeLa Cells , Humans , Mitosis , Nuclear Envelope/chemistry , Nuclear Envelope/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism , Phosphates/metabolism , Phosphopeptides/isolation & purification , Phosphopeptides/metabolism , Rats
2.
J Med Genet ; 58(11): 783-788, 2021 11.
Article in English | MEDLINE | ID: mdl-32938693

ABSTRACT

BACKGROUND: Triploidy is one of the most common chromosome abnormalities affecting human gestation and accounts for an important fraction of first-trimester miscarriages. Triploidy has been demonstrated in a few cases of recurrent pregnancy loss (RPL) but its molecular mechanisms are unknown. This study aims to identify the genetic cause of RPL associated with fetus triploidy. METHODS: We investigated genomic imprinting, genotyped sequence-tagged site (STS) markers and performed exome sequencing in a family including two sisters with RPL. Moreover, we evaluated oocyte maturation in vivo and in vitro and effect of the candidate protein variant in silico. RESULTS: While features of hydatidiform mole were excluded, the presence of triploidy of maternal origin was demonstrated in the fetuses. Oocyte maturation was deficient and all the maternally inherited pericentromeric STS alleles were homozygous in the fetuses. A deleterious missense variant (p.V1251D) of the cyclin B3 gene (CCNB3) affecting a residue conserved in placental mammals and located in a region that can interact with the cyclin-dependent kinase 1 or cyclin-dependent kinase 2 cosegregated in homozygosity with RPL. CONCLUSION: Here, we report a family in which a damaging variant in cyclin B3 is associated with the failure of oocyte meiosis II and recurrent fetus triploidy, implicating a rationale for CCNB3 testing in RPL.


Subject(s)
Abortion, Habitual/genetics , Cyclin B/genetics , Triploidy , Cyclin B/chemistry , Female , Humans , Meiosis/genetics , Oocytes/physiology , Pregnancy , Exome Sequencing
3.
Biochim Biophys Acta Gene Regul Mech ; 1863(7): 194549, 2020 07.
Article in English | MEDLINE | ID: mdl-32275964

ABSTRACT

The Cyclin B1 gene encodes a G2/M cyclin that is deregulated in various human cancers, however, the transcriptional regulation of this gene is incompletely understood. The E2F and retinoblastoma family of proteins are involved in this gene's regulation, but there is disagreement on which of the E2F and retinoblastoma proteins interact with the promoter to regulate this gene. Here, we dissect the promoter region of the Drosophila CycB gene, and study the role of Rbf and E2F factors in its regulation. This gene exhibits remarkable features that distinguish it from G1/S regulated promoters, such as PCNA. The promoter is comprised of modular elements with dedicated repressor and activator functions, including a segment spanning the first intron that interferes with a 5' activator element. A highly active minimal promoter (-464, +100) is repressed by the Rbf1 retinoblastoma protein, but much more potently repressed by the Rbf2 protein, which has been linked in other studies to control of cell growth genes. Unlike many other cell-cycle genes, which are activated by E2F1 and repressed by E2F2, CycB is potently activated by E2F2, and repressed by E2F1. Although the bulk of Rbf binding is associated with a region 5' of the core promoter, E2F and retinoblastoma proteins functionally interact with the basal promoter region, in part through a conserved E2F site at -80 bp. The specific regulatory requirements of this late cell cycle promoter appear to be linked to the unique activities of E2F and retinoblastoma family members acting on a complex cis-regulatory circuit.


Subject(s)
Cyclin B/genetics , Drosophila Proteins/genetics , Promoter Regions, Genetic , Transcriptional Activation , Animals , Cell Line , Conserved Sequence , Cyclin B/chemistry , Cyclin B/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster , Introns , Protein Binding , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Lab Chip ; 19(20): 3499-3511, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31544194

ABSTRACT

Cell-free Xenopus egg extract is a widely used and biochemically tractable model system that allows recapitulation and elucidation of fundamental cellular processes. Recently, the introduction of microfluidic extract manipulation has enabled compartmentalization of bulk extract and a newfound ability to study organelles on length scales that recapitulate key features of cellular morphology. While the microfluidic confinement of extracts has produced a compelling platform for the in vitro study of cell processes at physiologically-relevant length scales, it also imposes experimental limitations by restricting dynamic control over extract properties. Here, we introduce photodegradable polyethylene glycol (PEG) hydrogels as a vehicle to passively and selectively manipulate extract composition through the release of proteins encapsulated within the hydrogel matrix. Photopatterned PEG hydrogels, passive to both extract and encapsulated proteins, serve as protein depots within microfluidic channels, which are subsequently flooded with extract. Illumination by ultraviolet light (UV) degrades the hydrogel structures and releases encapsulated protein. We show that an engineered fluorescent protein with a nuclear localization signal (GST-GFP-NLS) retains its ability to localize within nearby nuclei following UV-induced release from hydrogel structures. When diffusion is considered, the kinetics of nuclear accumulation are similar to those in experiments utilizing conventional, bulk fluid handling. Similarly, the release of recombinant cyclin B Δ90, a mutant form of the master cell cycle regulator cyclin B which lacks the canonical destruction box, was able to induce the expected cell cycle transition from interphase to mitosis. This transition was confirmed by the observation of nuclear envelope breakdown (NEBD), a phenomenological hallmark of mitosis, and the induction of mitosis-specific biochemical markers. This approach to extract manipulation presents a versatile and customizable route to regulating the spatial and temporal dynamics of cellular events in microfluidically confined cell-free extracts.


Subject(s)
Microfluidics/methods , Mitosis , Ultraviolet Rays , Xenopus laevis/growth & development , Animals , Cell Nucleus/metabolism , Cyclin B/chemistry , Cyclin B/metabolism , Hydrogels/chemistry , Hydrogels/metabolism , Mitosis/drug effects , Mitosis/radiation effects , Oocytes/cytology , Oocytes/drug effects , Polyethylene Glycols/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Xenopus laevis/metabolism
5.
Chromosoma ; 128(3): 473-487, 2019 09.
Article in English | MEDLINE | ID: mdl-31446450

ABSTRACT

Cyclins, as regulatory partners of cyclin-dependent kinases (CDKs), control the switch-like cell cycle transitions that orchestrate orderly duplication and segregation of genomes. Compared to mitosis, relatively little is known about how cyclin-CDK complexes control meiosis, the specialized cell division that generates gametes for sexual production. Mouse cyclin B3 was previously shown to have expression restricted to the beginning of meiosis, making it a candidate to regulate meiotic events. Indeed, female mice lacking cyclin B3 are sterile because oocytes arrest at the metaphase-to-anaphase transition of meiosis I. However, whether cyclin B3 functions during spermatogenesis was untested. Here, we found that males lacking cyclin B3 are fertile and show no detectable defects in spermatogenesis based on histological analysis of seminiferous tubules. Cytological analysis further showed no detectable defects in homologous chromosome synapsis or meiotic progression, and suggested that recombination is initiated and completed efficiently. Moreover, absence of cyclin B3 did not exacerbate previously described meiotic defects in mutants deficient for cyclin E2, suggesting a lack of redundancy between these cyclins. Thus, unlike in females, cyclin B3 is not essential for meiosis in males despite its prominent meiosis-specific expression.


Subject(s)
Cyclin B/genetics , Spermatogenesis/genetics , Alleles , Amino Acid Sequence , Animals , Cyclin B/chemistry , Cyclin B/metabolism , Gene Editing , Gene Expression , Immunohistochemistry , Male , Meiosis , Metaphase/genetics , Mice , Prophase/genetics , Protein Domains , Recombination, Genetic
6.
Nature ; 532(7599): 398-401, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27074503

ABSTRACT

USP14 is a major regulator of the proteasome and one of three proteasome-associated deubiquitinating enzymes. Its effects on protein turnover are substrate-specific, for unknown reasons. We report that USP14 shows a marked preference for ubiquitin-cyclin B conjugates that carry more than one ubiquitin modification or chain. This specificity is conserved from yeast to humans and is independent of chain linkage type. USP14 has been thought to cleave single ubiquitin groups from the distal tip of a chain, but we find that it removes chains from cyclin B en bloc, proceeding until a single chain remains. The suppression of degradation by USP14's catalytic activity reflects its capacity to act on a millisecond time scale, before the proteasome can initiate degradation of the substrate. In addition, single-molecule studies showed that the dwell time of ubiquitin conjugates at the proteasome was reduced by USP14-dependent deubiquitination. In summary, the specificity of the proteasome can be regulated by rapid ubiquitin chain removal, which resolves substrates based on a novel aspect of ubiquitin conjugate architecture.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination , Biocatalysis , Cyclin B/chemistry , Cyclin B/metabolism , Humans , Kinetics , Models, Molecular , Proteolysis , Substrate Specificity , Ubiquitin/metabolism , Yeasts/enzymology
7.
Methods Mol Biol ; 1342: 201-8, 2016.
Article in English | MEDLINE | ID: mdl-26254925

ABSTRACT

Tightly controlled degradation of specific regulatory proteins is crucial for transitioning to the next cell cycle phase, ensuring precise DNA replication and an equal distribution of chromosomes to provide genomic stability and avoid tumorigenesis. To study mitotic control at the metaphase-to-anaphase transition, a histone H2-GFP-based reporter system was established, allowing simultaneous monitoring of the alignment of mitotic chromosomes and cyclin B proteolysis. To depict the proteolytic profile, a chimeric cyclin B-SNAP reporter molecule that can be labeled with a fluorochrome-carrying SNAP substrate was generated for measurement of the decline in fluorescence intensity via live-cell imaging. This reporter system can be adapted for other cell cycle oscillatory proteins.


Subject(s)
Cyclin B/chemistry , Cyclin B/metabolism , Fluorescent Dyes/chemistry , Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Line, Tumor , Humans , M Phase Cell Cycle Checkpoints , Proteolysis , Spectrometry, Fluorescence
8.
Int J Mol Sci ; 16(5): 9314-40, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25918937

ABSTRACT

Cyclin-dependent kinase 2 (CDK2) is a crucial regulator of the eukaryotic cell cycle. However it is well established that monomeric CDK2 lacks regulatory activity, which needs to be aroused by its positive regulators, cyclins E and A, or be phosphorylated on the catalytic segment. Interestingly, these activation steps bring some dynamic changes on the 3D-structure of the kinase, especially the activation segment. Until now, in the monomeric CDK2 structure, three binding sites have been reported, including the adenosine triphosphate (ATP) binding site (Site I) and two non-competitive binding sites (Site II and III). In addition, when the kinase is subjected to the cyclin binding process, the resulting structural changes give rise to a variation of the ATP binding site, thus generating an allosteric binding site (Site IV). All the four sites are demonstrated as being targeted by corresponding inhibitors, as is illustrated by the allosteric binding one which is targeted by inhibitor ANS (fluorophore 8-anilino-1-naphthalene sulfonate). In the present work, the binding mechanisms and their fluctuations during the activation process attract our attention. Therefore, we carry out corresponding studies on the structural characterization of CDK2, which are expected to facilitate the understanding of the molecular mechanisms of kinase proteins. Besides, the binding mechanisms of CDK2 with its relevant inhibitors, as well as the changes of binding mechanisms following conformational variations of CDK2, are summarized and compared. The summary of the conformational characteristics and ligand binding mechanisms of CDK2 in the present work will improve our understanding of the molecular mechanisms regulating the bioactivities of CDK2.


Subject(s)
Cyclin-Dependent Kinase 2/chemistry , Ligands , Adenosine Triphosphate/chemistry , Allosteric Site , Amino Acid Sequence , Binding Sites , Binding, Competitive , Catalysis , Clinical Trials as Topic , Cyclin A/chemistry , Cyclin B/chemistry , Cyclin E/chemistry , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Genetic Variation , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Structure, Tertiary
9.
Nat Commun ; 6: 6769, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25864384

ABSTRACT

CDK1 is the only essential cell cycle CDK in human cells and is required for successful completion of M-phase. It is the founding member of the CDK family and is conserved across all eukaryotes. Here we report the crystal structures of complexes of CDK1-Cks1 and CDK1-cyclin B-Cks2. These structures confirm the conserved nature of the inactive monomeric CDK fold and its ability to be remodelled by cyclin binding. Relative to CDK2-cyclin A, CDK1-cyclin B is less thermally stable, has a smaller interfacial surface, is more susceptible to activation segment dephosphorylation and shows differences in the substrate sequence features that determine activity. Both CDK1 and CDK2 are potential cancer targets for which selective compounds are required. We also describe the first structure of CDK1 bound to a potent ATP-competitive inhibitor and identify aspects of CDK1 structure and plasticity that might be exploited to develop CDK1-selective inhibitors.


Subject(s)
CDC2-CDC28 Kinases/chemistry , Carrier Proteins/chemistry , Cell Cycle Proteins/chemistry , Cyclin B/chemistry , Cyclin-Dependent Kinases/chemistry , Recombinant Fusion Proteins/chemistry , Adenosine Triphosphate/chemistry , Animals , Binding, Competitive , CDC2 Protein Kinase , CDC2-CDC28 Kinases/genetics , Carrier Proteins/genetics , Cattle , Cell Cycle Proteins/genetics , Conserved Sequence , Crystallography, X-Ray , Cyclin A/chemistry , Cyclin A/genetics , Cyclin B/genetics , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Gene Expression , Humans , Kinetics , Models, Molecular , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Substrate Specificity
10.
PLoS One ; 9(1): e85343, 2014.
Article in English | MEDLINE | ID: mdl-24404204

ABSTRACT

During embryonic development of Artemia sinica, environmental stresses induce the embryo diapause phenomenon, required to resist apoptosis and regulate cell cycle activity. The small ubiquitin-related modifier-1 (SUMO), a reversible post-translational protein modifier, plays an important role in embryo development. SUMO regulates multiple cellular processes, including development and other biological processes. The molecular mechanism of diapause, diapause termination and the role of As-sumo-1 in this processes and in early embryo development of Artemia sinica still remains unknown. In this study, the complete cDNA sequences of the sumo-1 homolog, sumo ligase homolog, caspase-1 homolog and cyclin B homolog from Artemia sinica were cloned. The mRNA expression patterns of As-sumo-1, sumo ligase, caspase-1, cyclin B and the location of As-sumo-1 were investigated. SUMO-1, p53, Mdm2, Caspase-1, Cyclin B and Cyclin E proteins were analyzed during different developmental stages of the embryo of A. sinica. Small interfering RNA (siRNA) was used to verify the function of sumo-1 in A. sinica. The full-length cDNA of As-sumo-1 was 476 bp, encoding a 92 amino acid protein. The As-caspases-1 cDNA was 966 bp, encoding a 245 amino-acid protein. The As-sumo ligase cDNA was 1556 bp encoding, a 343 amino acid protein, and the cyclin B cDNA was 739 bp, encoding a 133 amino acid protein. The expressions of As-sumo-1, As-caspase-1 and As-cyclin B were highest at the 10 h stage of embryonic development, and As-sumo ligase showed its highest expression at 0 h. The expression of As-SUMO-1 showed no tissue or organ specificity. Western blotting showed high expression of As-SUMO-1, p53, Mdm2, Caspase-1, Cyclin B and Cyclin E at the 10 h stage. The siRNA caused abnormal development of the embryo, with increased malformation and mortality. As-SUMO-1 is a crucial regulation and modification protein resumption of embryonic diapause and early embryo development of A. sinica.


Subject(s)
Artemia/embryology , Artemia/genetics , SUMO-1 Protein/genetics , Amino Acid Sequence , Animals , Artemia/classification , Artemia/metabolism , Base Sequence , Caspase 1/chemistry , Caspase 1/genetics , Caspase 1/metabolism , Cloning, Molecular , Computational Biology , Cyclin B/chemistry , Cyclin B/genetics , Cyclin B/metabolism , Gene Expression Regulation, Developmental , Molecular Sequence Data , Phylogeny , RNA Interference , SUMO-1 Protein/chemistry , SUMO-1 Protein/metabolism , Sequence Alignment
11.
J Vis Exp ; (67): e4239, 2012 Sep 17.
Article in English | MEDLINE | ID: mdl-23007658

ABSTRACT

Equal distribution of chromosomes between the two daughter cells during cell division is a prerequisite for guaranteeing genetic stability. Inaccuracies during chromosome separation are a hallmark of malignancy and associated with progressive disease. The spindle assembly checkpoint (SAC) is a mitotic surveillance mechanism that holds back cells at metaphase until every single chromosome has established a stable bipolar attachment to the mitotic spindle. The SAC exerts its function by interference with the activating APC/C subunit Cdc20 to block proteolysis of securin and cyclin B and thus chromosome separation and mitotic exit. Improper attachment of chromosomes prevents silencing of SAC signaling and causes continued inhibition of APC/C(Cdc20) until the problem is solved to avoid chromosome missegregation, aneuploidy and malignant growths. Most studies that addressed the influence of improper chromosomal attachment on APC/C-dependent proteolysis took advantage of spindle disruption using depolymerizing or microtubule-stabilizing drugs to interfere with chromosomal attachment to microtubules. Since interference with microtubule kinetics can affect the transport and localization of critical regulators, these procedures bear a risk of inducing artificial effects. To study how the SAC interferes with APC/C-dependent proteolysis of cyclin B during mitosis in unperturbed cell populations, we established a histone H2-GFP-based system which allowed the simultaneous monitoring of metaphase alignment of mitotic chromosomes and proteolysis of cyclin B. To depict proteolytic profiles, we generated a chimeric cyclin B reporter molecule with a C-terminal SNAP moiety (Figure 1). In a self-labeling reaction, the SNAP-moiety is able to form covalent bonds with alkylguanine-carriers (SNAP substrate) (Figure 1). SNAP substrate molecules are readily available and carry a broad spectrum of different fluorochromes. Chimeric cyclin B-SNAP molecules become labeled upon addition of the membrane-permeable SNAP substrate to the growth medium (Figure 1). Following the labeling reaction, the cyclin B-SNAP fluorescence intensity drops in a pulse-chase reaction-like manner and fluorescence intensities reflect levels of cyclin B degradation (Figure 1). Our system facilitates the monitoring of mitotic APC/C-dependent proteolysis in large numbers of cells (or several cell populations) in parallel. Thereby, the system may be a valuable tool to identify agents/small molecules that are able to interfere with proteolytic activity at the metaphase to anaphase transition. Moreover, as synthesis of cyclin B during mitosis has recently been suggested as an important mechanism in fostering a mitotic block in mice and humans by keeping cyclin B expression levels stable, this system enabled us to analyze cyclin B proteolysis as one element of a balanced equilibrium.


Subject(s)
Cyclin B/metabolism , Microscopy, Fluorescence/methods , Single-Cell Analysis/methods , Bone Neoplasms/chemistry , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cyclin B/chemistry , Fluorescent Dyes/chemistry , Humans , Osteosarcoma/chemistry , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proteolysis , Recombinant Fusion Proteins/chemistry , SNARE Proteins/chemistry , Spindle Apparatus/chemistry , Spindle Apparatus/metabolism
12.
Article in English | MEDLINE | ID: mdl-22841647

ABSTRACT

The maturation promoting factor (MPF) is a key regulator of controlling G2/M phase transition in the meiotic maturation of oocyte and spermatocyte in animals, which is a complex of CDC2 (CDK1) and cyclin B. To better understand the molecular mechanism of oocyte and spermatocyte maturation in mud crab (Scylla paramamosain), the full length cDNA of cdc2 (Sp-cdc2) and cyclin B (Sp-cyclin B) were cloned and characterized. The full length cDNA of Sp-cdc2 gene is of 1593 bp encoding a protein of 299 amino acids. Real-time quantitative PCR analysis revealed that the expression level of Sp-cdc2 in the ovary was higher than in other tissues (P<0.01); and its expression level was not significantly different in different stages of ovary development (P>0.05), meanwhile there was higher expression in T3 stage than in T1 and T2 stages (P<0.05). The full length cDNA of Sp-cyclin B is 1492 bp encoding a protein of 391 amino acids. The real-time PCR results showed that its expression level in the ovary was the highest in all examined tissues (P<0.01), and the gonad expression level in O5 stage was significantly higher than in previous 4 stages and the testis (P<0.05), and was also significantly higher in T2 stage than in T1 stage (P<0.05). In situ hybridization analysis showed that the expressions of Sp-cdc2 and Sp-cyclin B transcripts were presented in similar distribution patterns in different developing stages of ovary and testis. The positive signals of Sp-cdc2 and Sp-cyclin B mRNA were detected in the oocytoplasm of oogonia and pre-vitellogenic and primary vitellogenic oocytes, while these two genes had higher expression level in the spermatid and secondary spermatocyte following primary spermatocyte. These results suggested that Sp-cdc2 and Sp-cyclin B may play essential roles in the oogenesis and spermatogenesis of the crab.


Subject(s)
Brachyura/growth & development , Brachyura/genetics , CDC2 Protein Kinase/genetics , Cyclin B/genetics , Gene Expression Regulation, Developmental , Oogenesis/genetics , Spermatogenesis/genetics , Amino Acid Sequence , Animals , Base Sequence , Brachyura/enzymology , Brachyura/metabolism , CDC2 Protein Kinase/chemistry , CDC2 Protein Kinase/metabolism , Cloning, Molecular , Cyclin B/chemistry , Cyclin B/metabolism , DNA, Complementary/genetics , Female , Male , Molecular Sequence Data , Oocytes/cytology , Oocytes/metabolism , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Spermatozoa/cytology , Spermatozoa/metabolism
13.
Philos Trans R Soc Lond B Biol Sci ; 366(1584): 3584-94, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22084385

ABSTRACT

Activation of the cyclin-dependent kinase (Cdk1) cyclin B (CycB) complex (Cdk1:CycB) in mitosis brings about a remarkable extent of protein phosphorylation. Cdk1:CycB activation is switch-like, controlled by two auto-amplification loops--Cdk1:CycB activates its activating phosphatase, Cdc25, and inhibits its inhibiting kinase, Wee1. Recent experimental evidence suggests that parallel to Cdk1:CycB activation during mitosis, there is inhibition of its counteracting phosphatase activity. We argue that the downregulation of the phosphatase is not just a simple latch that suppresses futile cycles of phosphorylation/dephosphorylation during mitosis. Instead, we propose that phosphatase regulation creates coherent feed-forward loops and adds extra amplification loops to the Cdk1:CycB regulatory network, thus forming an integral part of the mitotic switch. These network motifs further strengthen the bistable characteristic of the mitotic switch, which is based on the antagonistic interaction of two groups of proteins: M-phase promoting factors (Cdk1:CycB, Cdc25, Greatwall and Endosulfine/Arpp19) and interphase promoting factors (Wee1, PP2A-B55 and a Greatwall counteracting phosphatase, probably PP1). The bistable character of the switch implies the existence of a CycB threshold for entry into mitosis. The end of G2 phase is determined by the point where CycB level crosses the CycB threshold for Cdk1 activation.


Subject(s)
CDC2 Protein Kinase/chemistry , Cell Cycle Checkpoints , Cyclin B/chemistry , Mitosis , Animals , Enzyme Activation , Fungal Proteins/chemistry , Interphase , Phosphorylation , Yeasts/chemistry , Yeasts/cytology , cdc25 Phosphatases/chemistry
14.
Philos Trans R Soc Lond B Biol Sci ; 366(1584): 3625-34, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22084388

ABSTRACT

'…in Italy, for thirty years under the Borgias, they had warfare, terror, murder and bloodshed, but they produced Michelangelo, Leonardo da Vinci and the Renaissance. In Switzerland, they had brotherly love, they had five hundred years of democracy and peace-and what did that produce? The cuckoo clock'. Orson Welles as Harry Lime: The Third Man. Orson Welles might have been a little unfair on the Swiss, after all cuckoo clocks were developed in the Schwartzwald, but, more importantly, Swiss democracy gives remarkably stable government with considerable decision-making at the local level. The alternative is the battling city-states of Renaissance Italy: culturally rich but chaotic at a higher level of organization. As our understanding of the cell cycle improves, it appears that the cell is organized more along the lines of Switzerland than Renaissance Italy, and one major challenge is to determine how local decisions are made and coordinated to produce the robust cell cycle mechanisms that we observe in the cell as a whole.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle , DNA Replication , Anaphase-Promoting Complex-Cyclosome , Animals , Centrosome/chemistry , Cyclin B/chemistry , Cyclin-Dependent Kinases/chemistry , Fungal Proteins/chemistry , Humans , Kinetochores/chemistry , M Phase Cell Cycle Checkpoints , Signal Transduction , Ubiquitin-Protein Ligase Complexes/chemistry , Yeasts/chemistry , cdc25 Phosphatases/chemistry
15.
Philos Trans R Soc Lond B Biol Sci ; 366(1584): 3595-604, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22084386

ABSTRACT

The spindle assembly checkpoint controls cell cycle progression during mitosis, synchronizing it with the attachment of chromosomes to spindle microtubules. After the discovery of the mitotic arrest deficient (MAD) and budding uninhibited by benzymidazole (BUB) genes as crucial checkpoint components in 1991, the second decade of checkpoint studies (2001-2010) witnessed crucial advances in the elucidation of the mechanism through which the checkpoint effector, the mitotic checkpoint complex, targets the anaphase-promoting complex (APC/C) to prevent progression into anaphase. Concomitantly, the discovery that the Ndc80 complex and other components of the microtubule-binding interface of kinetochores are essential for the checkpoint response finally asserted that kinetochores are crucial for the checkpoint response. Nevertheless, the relationship between kinetochores and checkpoint control remains poorly understood. Crucial advances in this area in the third decade of checkpoint studies (2011-2020) are likely to be brought about by the characterization of the mechanism of kinetochore recruitment, activation and inactivation of checkpoint proteins, which remains elusive for the majority of checkpoint components. Here, we take a molecular view on the main challenges hampering this task.


Subject(s)
Cell Cycle Proteins/chemistry , Kinetochores/chemistry , M Phase Cell Cycle Checkpoints , Animals , Aurora Kinases , Binding Sites , Cyclin B/chemistry , Enzyme Activation , Humans , Multiprotein Complexes/chemistry , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Signal Transduction , Spindle Apparatus
16.
Philos Trans R Soc Lond B Biol Sci ; 366(1584): 3638-52, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22084390

ABSTRACT

Translational regulation contributes to the control of archetypal and specialized cell cycles, such as the meiotic and early embryonic cycles. Late meiosis and early embryogenesis unfold in the absence of transcription, so they particularly rely on translational repression and activation of stored maternal mRNAs. Here, we present examples of cell cycle regulators that are translationally controlled during different cell cycle and developmental transitions in model organisms ranging from yeast to mouse. Our focus also is on the RNA-binding proteins that affect cell cycle progression by recognizing special features in untranslated regions of mRNAs. Recent research highlights the significance of the cytoplasmic polyadenylation element-binding protein (CPEB). CPEB determines polyadenylation status, and consequently translational efficiency, of its target mRNAs in both transcriptionally active somatic cells as well as in transcriptionally silent mature Xenopus oocytes and early embryos. We discuss the role of CPEB in mediating the translational timing and in some cases spindle-localized translation of critical regulators of Xenopus oogenesis and early embryogenesis. We conclude by outlining potential directions and approaches that may provide further insights into the translational control of the cell cycle.


Subject(s)
Cell Cycle , Protein Biosynthesis , Animals , Cyclin B/chemistry , Embryonic Development , Male , Meiosis , Mitosis , Oocytes/chemistry , Oocytes/growth & development , Polyadenylation , RNA-Binding Proteins/chemistry , Repressor Proteins/chemistry , Spermatocytes/chemistry , Spermatocytes/growth & development , Transcription Factors/chemistry , Xenopus/embryology , Xenopus/growth & development , Xenopus Proteins/chemistry , mRNA Cleavage and Polyadenylation Factors/chemistry
17.
Cell Cycle ; 10(22): 3957-63, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22071690

ABSTRACT

The origin recognition complex (ORC) is an essential component of the pre-replicative complex (pre-RC) that binds to replication origins for licensing. Levels of the largest ORC subunit, ORC1, oscillate during the mitotic cell cycle and regulate origin usage. In Drosophila, ORC1 levels increase at the G(1)/S transition following E2F-dependent transcriptional activation, remain high until the end of M phase and then decrease at the M/G(1) transition when ORC1 is targeted for proteolysis by the anaphase-promoting complex (APC). A function, if any, for Drosophila ORC1 after S phase has not been described. Here, we determined the role of ORC1 at stages outside S phase by generating ORC1 derivatives with a modified ORC1 degradation box (the O-box) and examining the effects in vivo. These modifications either stabilized ORC1 by mutating the O-box (ORC1(Omut)) so that it is no longer targeted by APC or changed its degradation profile by replacing the O-box with the D-box of human cyclin B (ORC1(O→D)), so that degradation would occur earlier. We determined the distribution and tested the function of these ORC1 derivatives in an orc1 mutant background so that only the mutated protein was expressed. Stable version of ORC1, ORC1 (Omut), showed no effects on cell cycle progression; however, ORC1(O→D), which is degraded early at the G(2)/M transition, led to a higher frequency of M-phase cells but not S-phase cells. Taken together, our results indicate the timing of ORC1 degradation is required for timely progression in M phase.


Subject(s)
DNA Replication , Drosophila Proteins/physiology , Drosophila/genetics , Origin Recognition Complex/physiology , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Division , Cyclin B/chemistry , Cyclin B/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Humans , Origin Recognition Complex/chemistry , Origin Recognition Complex/genetics , Proteolysis , S Phase , Ubiquitin-Protein Ligase Complexes/physiology
18.
Proc Natl Acad Sci U S A ; 106(41): 17337-42, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19805136

ABSTRACT

The first telomerase cofactor identified was the budding yeast protein Est1, which is conserved through humans. While it is evident that Est1 is required for telomere DNA maintenance, understanding its mechanistic contributions to telomerase regulation has been limited. In vitro, the primary effect of Est1 is to activate telomerase-mediated DNA extension. Although Est1 displayed specific DNA and RNA binding, neither activity contributed significantly to telomerase stimulation. Rather Est1 mediated telomerase upregulation through direct contacts with the reverse transcriptase subunit. In addition to intrinsic Est1 functions, we found that Est1 cooperatively activated telomerase in conjunction with Cdc13 and that the combinatorial effect was dependent upon a known salt-bridge interaction between Est1 (K444) and Cdc13 (E252). Our studies provide insights into the molecular events used to control the enzymatic activity of the telomerase holoenzyme.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Telomerase/metabolism , Animals , Binding Sites , Cyclin B/chemistry , Cyclin B/metabolism , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Enzyme Activation , Humans , Nucleic Acid Hybridization , RNA/chemistry , RNA/metabolism , RNA, Fungal/chemistry , RNA, Fungal/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/isolation & purification , Telomerase/chemistry , Telomerase/isolation & purification
19.
J Biol Chem ; 284(42): 28775-82, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19687009

ABSTRACT

CLIP-170, the founding member of microtubule "plus ends tracking" proteins, is involved in many critical microtubule-related functions, including recruitment of dynactin to the microtubule plus ends and formation of kinetochore-microtubule attachments during metaphase. Although it has been reported that CLIP-170 is a phosphoprotein, neither have individual phosphorylation sites been identified nor have the associated kinases been extensively studied. Herein, we identify Cdc2 as a kinase that phosphorylates CLIP-170. We show that Cdc2 interacts with CLIP-170 mediating its phosphorylation on Thr(287) in vivo. Significantly, expression of CLIP-170 with a threonine 287 to alanine substitution (T287A) results in its mislocalization, accumulation of Plk1 and cyclin B, and block of the G2/M transition. Finally, we found that depletion of CLIP-170 leads to centrosome reduplication and that Cdc2 phosphorylation of CLIP-170 is required for the process. These results demonstrate that Cdc2-mediated phosphorylation of CLIP-170 is essential for the normal function of this protein during cell cycle progression.


Subject(s)
Centrosome/ultrastructure , Cyclin B/chemistry , Cyclin B/physiology , Microtubule-Associated Proteins/chemistry , Neoplasm Proteins/chemistry , Animals , CDC2 Protein Kinase , Cell Cycle , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinases , Humans , Microtubules/metabolism , Peptides/chemistry , Phenotype , Phosphorylation , Rats , Recombinant Proteins/chemistry , Tubulin/chemistry
20.
J Exp Bot ; 60(9): 2621-30, 2009.
Article in English | MEDLINE | ID: mdl-19414499

ABSTRACT

Cell cycle progression requires interaction between cyclin-dependent kinase B (CDKB) and cyclin B (CYCB). The seasonal expression patterns of the CDKB and CYCB homologues from Populus tomentosa Carr. were investigated, and effects of temperature and exogenous indole-3-acetic acid (IAA) on their expression were further studied in water culture experiments. Based on the differential responses of dormant cambium cells to exogenous IAA, four stages of cambium dormancy were confirmed for P. tomentosa: quiescence 1 (Q1), rest, quiescence 2-1 (Q2-1), and quiescence 2-2 (Q2-2). PtoCDKB and PtoCYCB transcripts were strongly expressed in the active phases, weakly in Q1, and almost undetectable from rest until late Q2-2. Climatic data analysis showed a correlation between daily air temperature and PtoCDKB and PtoCYCB expression patterns. Water culture experiments with temperature treatment further showed that a low temperature (4 degrees C) kept PtoCDKB and PtoCYCB transcripts at undetectable levels, while a warm temperature (25 degrees C) induced their expression in the cambium region. Meanwhile, water culture experiments with exogenous IAA treatment showed that induction of PtoCDKB and PtoCYCB transcription was independent of exogenous IAA. The results suggest that, in deciduous hardwood P. tomentosa growing in a temperate zone, the temperature in early spring is a vital environmental factor for cambium reactivation. The increasing temperature in early spring may induce CDKB and CYCB homologue transcription in the cambium region, which is necessary for cambium cell division.


Subject(s)
Cyclin B/genetics , Cyclin-Dependent Kinases/genetics , Meristem/genetics , Plant Proteins/genetics , Populus/genetics , Transcription, Genetic , Up-Regulation , Amino Acid Sequence , Cell Cycle , Cyclin B/chemistry , Cyclin B/metabolism , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Plant , Indoleacetic Acids/metabolism , Meristem/chemistry , Meristem/metabolism , Molecular Sequence Data , Plant Proteins/chemistry , Plant Proteins/metabolism , Populus/chemistry , Populus/metabolism , Sequence Alignment , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL