To see the other types of publications on this topic, follow the link: Polo kinases/Cdc5.

Journal articles on the topic 'Polo kinases/Cdc5'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Polo kinases/Cdc5.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Song, Sukgil, Tallessyn Z. Grenfell, Susan Garfield, Raymond L. Erikson, and Kyung S. Lee. "Essential Function of the Polo Box of Cdc5 in Subcellular Localization and Induction of Cytokinetic Structures." Molecular and Cellular Biology 20, no. 1 (January 1, 2000): 286–98. http://dx.doi.org/10.1128/mcb.20.1.286-298.2000.

Full text
Abstract:
ABSTRACT Members of the polo subfamily of protein kinases play pivotal roles in cell proliferation. In addition to the kinase domain, polo kinases have a strikingly conserved sequence in the noncatalytic C-terminal domain, termed the polo box. Here we show that the budding-yeast polo kinase Cdc5, when fused to green fluorescent protein and expressed under its endogenous promoter, localizes at spindle poles and the mother bud neck. Overexpression of Cdc5 can induce a class of cells with abnormally elongated buds in a polo box- and kinase activity-dependent manner. In addition to localizing at the spindle poles and cytokinetic neck filaments, Cdc5 induces and localizes to additional septin ring structures within the elongated buds. Without impairing kinase activity, conservative mutations in the polo box abolish the ability of Cdc5 to functionally complement the defect associated with a cdc5-1 temperature-sensitive mutation, to localize to the spindle poles and cytokinetic neck filaments, and to induce elongated cells with ectopic septin ring structures. Consistent with the polo box-dependent subcellular localization, the C-terminal domain of Cdc5, but not its polo box mutant, is sufficient for subcellular localization, and its overexpression appears to inhibit cytokinesis. These data provide evidence that the polo box is required to direct Cdc5 to specific subcellular locations and induce or organize cytokinetic structures.
APA, Harvard, Vancouver, ISO, and other styles
2

Hardy, C. F., and A. Pautz. "A novel role for Cdc5p in DNA replication." Molecular and Cellular Biology 16, no. 12 (December 1996): 6775–82. http://dx.doi.org/10.1128/mcb.16.12.6775.

Full text
Abstract:
DNA replication initiates from specific chromosomal sites called origins, and in the budding yeast Saccharomyces cerevisiae these sites are occupied by the origin recognition complex (ORC). Dbf4p is proposed to play a role in targeting the G1/S kinase Cdc7p to initiation complexes late in G1. We report that Dbf4p may also recruit Cdc5p to origin complexes. Cdc5p is a member of the Polo family of kinases that is required for the completion of mitosis. Cdc5p and Cdc7p each interact with a distinct domain of Dbf4p. cdc5-1 mutants have a plasmid maintenance defect that can be suppressed by the addition of multiple origins. cdc5-1 orc2-1 double mutants are synthetically lethal. Levels of Cdc5p were found to be cell cycle regulated and peaked in G2/M. These results suggest a role for Cdc5p and possibly Polo-like kinases at origin complexes.
APA, Harvard, Vancouver, ISO, and other styles
3

Park, Chong J., Jung-Eun Park, Tatiana S. Karpova, Nak-Kyun Soung, Li-Rong Yu, Sukgil Song, Kyung H. Lee, et al. "Requirement for the Budding Yeast Polo Kinase Cdc5 in Proper Microtubule Growth and Dynamics." Eukaryotic Cell 7, no. 3 (January 4, 2008): 444–53. http://dx.doi.org/10.1128/ec.00283-07.

Full text
Abstract:
ABSTRACT In many organisms, polo kinases appear to play multiple roles during M-phase progression. To provide new insights into the function of the budding yeast polo kinase Cdc5, we generated novel temperature-sensitive cdc5 mutants by mutagenizing the C-terminal noncatalytic polo box domain, a region that is critical for proper subcellular localization. One of these mutants, cdc5-11, exhibited a temperature-sensitive growth defect with an abnormal spindle morphology. Strikingly, provision of a moderate level of benomyl, a microtubule-depolymerizing drug, permitted cdc5-11 cells to grow significantly better than the isogenic CDC5 wild type in a FEAR (cdc Fourteen Early Anaphase Release)-independent manner. In addition, cdc5-11 required MAD2 for both cell growth and the benomyl-remedial phenotype. These results suggest that cdc5-11 is defective in proper spindle function. Consistent with this view, cdc5-11 exhibited abnormal spindle morphology, shorter spindle length, and delayed microtubule regrowth at the nonpermissive temperature. Overexpression of CDC5 moderately rescued the spc98-2 growth defect. Interestingly, both Cdc28 and Cdc5 were required for the proper modification of the spindle pole body components Nud1, Slk19, and Stu2 in vivo. They also phosphorylated these three proteins in vitro. Taken together, these observations suggest that concerted action of Cdc28 and Cdc5 on Nud1, Slk19, and Stu2 is important for proper spindle functions.
APA, Harvard, Vancouver, ISO, and other styles
4

Park, Chong Jin, Sukgil Song, Philip R. Lee, Wenying Shou, Raymond J. Deshaies, and Kyung S. Lee. "Loss of CDC5 Function in Saccharomyces cerevisiae Leads to Defects in Swe1p Regulation and Bfa1p/Bub2p-Independent Cytokinesis." Genetics 163, no. 1 (January 1, 2003): 21–33. http://dx.doi.org/10.1093/genetics/163.1.21.

Full text
Abstract:
Abstract In many organisms, polo kinases appear to play multiple roles during M-phase progression. To provide new insights into the function of budding yeast polo kinase Cdc5p, we generated novel temperature-sensitive cdc5 mutants by mutagenizing the C-terminal domain. Here we show that, at a semipermissive temperature, the cdc5-3 mutant exhibited a synergistic bud elongation and growth defect with loss of HSL1, a component important for normal G2/M transition. Loss of SWE1, which phosphorylates and inactivates the budding yeast Cdk1 homolog Cdc28p, suppressed the cdc5-3 hsl1Δ defect, suggesting that Cdc5p functions at a point upstream of Swe1p. In addition, the cdc5-4 and cdc5-7 mutants exhibited chained cell morphologies with shared cytoplasms between the connected cell bodies, indicating a cytokinetic defect. Close examination of these mutants revealed delayed septin assembly at the incipient bud site and loosely organized septin rings at the mother-bud neck. Components in the mitotic exit network (MEN) play important roles in normal cytokinesis. However, loss of BFA1 or BUB2, negative regulators of the MEN, failed to remedy the cytokinetic defect of these mutants, indicating that Cdc5p promotes cytokinesis independently of Bfa1p and Bub2p. Thus, Cdc5p contributes to the activation of the Swe1p-dependent Cdc28p/Clb pathway, normal septin function, and cytokinesis.
APA, Harvard, Vancouver, ISO, and other styles
5

Golsteyn, R. M., S. J. Schultz, J. Bartek, A. Ziemiecki, T. Ried, and E. A. Nigg. "Cell cycle analysis and chromosomal localization of human Plk1, a putative homologue of the mitotic kinases Drosophila polo and Saccharomyces cerevisiae Cdc5." Journal of Cell Science 107, no. 6 (June 1, 1994): 1509–17. http://dx.doi.org/10.1242/jcs.107.6.1509.

Full text
Abstract:
polo and CDC5 are two genes required for passage through mitosis in Drosophila melanogaster and Saccharomyces cerevisiae, respectively. Both genes encode structurally related protein kinases that have been implicated in regulating the function of the mitotic spindle. Here, we report the characterization of a human protein kinase that displays extensive sequence similarity to Drosophila polo and S. cerevisiae Cdc5; we refer to this kinase as Plk1 (for polo-like kinase 1). The largest open reading frame of the Plk1 cDNA encodes a protein of 68,254 daltons, and a protein of this size is detected by immunoblotting of HeLa cell extracts with monoclonal antibodies raised against the C-terminal part of Plk1 expressed in Escherichia coli. Northern blot analysis of RNA isolated from human cells and mouse tissues shows that a single Plk1 mRNA of 2.3 kb is highly expressed in tissues with a high mitotic index, consistent with a possible function of Plk1 in cell proliferation. The Plk1 gene maps to position p12 on chromosome 16, a locus for which no associations with neoplastic malignancies are known. The Plk1 protein levels and its distribution change during the cell cycle, in a manner consistent with a role of Plk1 in mitosis. Thus, like Drosophila polo and S. cerevisiae Cdc5, human Plk1 is likely to function in cell cycle progression.
APA, Harvard, Vancouver, ISO, and other styles
6

Höfken, Thomas, and Elmar Schiebel. "Novel regulation of mitotic exit by the Cdc42 effectors Gic1 and Gic2." Journal of Cell Biology 164, no. 2 (January 19, 2004): 219–31. http://dx.doi.org/10.1083/jcb.200309080.

Full text
Abstract:
The guanine nucleotide exchange factor Cdc24, the GTPase Cdc42, and the Cdc42 effectors Cla4 and Ste20, two p21-activated kinases, form a signal transduction cascade that promotes mitotic exit in yeast. We performed a genetic screen to identify components of this pathway. Two related bud cortex–associated Cdc42 effectors, Gic1 and Gic2, were obtained as factors that promoted mitotic exit independently of Ste20. The mitotic exit function of Gic1 was dependent on its activation by Cdc42 and on the release of Gic1 from the bud cortex. Gic proteins became essential for mitotic exit when activation of the mitotic exit network through Cdc5 polo kinase and the bud cortex protein Lte1 was impaired. The mitotic exit defect of cdc5-10 Δlte1 Δgic1 Δgic2 cells was rescued by inactivation of the inhibiting Bfa1-Bub2 GTPase-activating protein. Moreover, Gic1 bound directly to Bub2 and prevented binding of the GTPase Tem1 to Bub2. We propose that in anaphase the Cdc42-regulated Gic proteins trigger mitotic exit by interfering with Bfa1-Bub2 GTPase-activating protein function.
APA, Harvard, Vancouver, ISO, and other styles
7

Lee, K. S., and R. L. Erikson. "Plk is a functional homolog of Saccharomyces cerevisiae Cdc5, and elevated Plk activity induces multiple septation structures." Molecular and Cellular Biology 17, no. 6 (June 1997): 3408–17. http://dx.doi.org/10.1128/mcb.17.6.3408.

Full text
Abstract:
Plk is a mammalian serine/threonine protein kinase whose activity peaks at the onset of M phase. It is closely related to other mammalian kinases, Snk, Fnk, and Prk, as well as to Xenopus laevis Plx1, Drosophila melanogaster polo, Schizosaccharomyces pombe Plo1, and Saccharomyces cerevisiae Cdc5. The M phase of the cell cycle is a highly coordinated process which insures the equipartition of genetic and cellular materials during cell division. To enable understanding of the function of Plk during M phase progression, various Plk mutants were generated and expressed in Sf9 cells and budding yeast. In vitro kinase assays with Plk immunoprecipitates prepared from Sf9 cells indicate that Glu206 and Thr210 play equally important roles for Plk activity and that replacement of Thr210 with a negatively charged residue elevates Plk specific activity. Ectopic expression of wild-type Plk (Plk WT) complements the cell division defect associated with the cdc5-1 mutation in S. cerevisiae. The degree of complementation correlates closely with the Plk activity measured in vitro, as it is enhanced by a mutationally activated Plk, T210D, but is not observed with the inactive forms K82M, D194N, and D194R. In a CDC5 wild-type background, expression of Plk WT or T210D, but not of inactive forms, induced a sharp accumulation of cells in G1. Consistent with elevated Plk activity, this phenomenon was enhanced by the C-terminally deleted forms WT deltaC and T210D deltaC. Expression of T210D also induced a class of cells with unusually elongated buds which developed multiple septal structures. This was not observed with the C-terminally deleted form T210D deltaC, however. It appears that the C terminus of Plk is not required for the observed cell cycle influence but may be important for polarized cell growth and septal structure formation.
APA, Harvard, Vancouver, ISO, and other styles
8

Park, Chong J., Sukgil Song, Thomas H. Giddings, Hyeon-Su Ro, Krisada Sakchaisri, Jung-Eun Park, Yeon-Sun Seong, Mark Winey, and Kyung S. Lee. "Requirement for Bbp1p in the Proper Mitotic Functions of Cdc5p in Saccharomyces cerevisiae." Molecular Biology of the Cell 15, no. 4 (April 2004): 1711–23. http://dx.doi.org/10.1091/mbc.e03-07-0461.

Full text
Abstract:
The polo-box domain of the budding yeast polo kinase Cdc5p plays an essential role for targeting the catalytic activity of Cdc5p to spindle pole bodies (SPBs) and cytokinetic neck-filaments. Here, we report the isolation of Bbp1p as a polo-box interacting protein by a yeast two-hybrid screen. Bbp1p localizes to the periphery of the central plaque of the SPB and plays an important role in SPB duplication. Similarly, Cdc5p localized to the cytoplasmic periphery of the SPB. In vitro binding studies showed that Cdc5p interacted with the N-terminal domain of Bbp1p (Bbp1pΔC), but apparently not with Mps2p, a component shown to form a stable complex with Bbp1p. In addition, Bbp1p, but likely not Mps2p, was required for proper localization of Cdc5p to the SPB. The C-terminal coiled-coil domain of Bbp1p (Bbp1p243–385), which is crucial for both the homodimerization and the SPB localization, could target the localization-defective Cdc5pΔC to the SPB and induce the release of Cdc14p from the nucleolus. Consistent with this observation, expression of CDC5ΔC-BBP1243–385 under CDC5 promoter control partially complemented the cdc5Δ defect. These data suggest that Bbp1pΔC interacts with the polo-box domain of Cdc5p, and this interaction is critical for the subcellular localization and mitotic functions of Cdc5p.
APA, Harvard, Vancouver, ISO, and other styles
9

Park, Jung-Eun, Chong J. Park, Krisada Sakchaisri, Tatiana Karpova, Satoshi Asano, James McNally, Yangil Sunwoo, Sun-Hee Leem, and Kyung S. Lee. "Novel Functional Dissection of the Localization-Specific Roles of Budding Yeast Polo Kinase Cdc5p." Molecular and Cellular Biology 24, no. 22 (November 15, 2004): 9873–86. http://dx.doi.org/10.1128/mcb.24.22.9873-9886.2004.

Full text
Abstract:
ABSTRACT Budding yeast polo kinase Cdc5p localizes to the spindle pole body (SPB) and to the bud-neck and plays multiple roles during M-phase progression. To dissect localization-specific mitotic functions of Cdc5p, we tethered a localization-defective N-terminal kinase domain of Cdc5p (Cdc5pΔC) to the SPB or to the bud-neck with components specifically localizing to one of these sites and characterized these mutants in a cdc5Δ background. Characterization of a viable, SPB-localizing, CDC5ΔC-CNM67 mutant revealed that it is defective in timely degradation of Swe1p, a negative regulator of Cdc28p. Loss of BFA1, a negative regulator of mitotic exit, rescued the lethality of a neck-localizing CDC5ΔC-CDC12 or CDC5ΔC-CDC3 mutant but yielded severe defects in cytokinesis. These data suggest that the SPB-associated Cdc5p activity is critical for both mitotic exit and cytokinesis, whereas the bud neck-localized Cdc5p is required for proper Swe1p regulation. Interestingly, a cdc5Δ bfa1Δ swe1Δ triple mutant is viable but grows slowly, whereas cdc5Δ cells bearing both CDC5ΔC-CNM67 and CDC5ΔC-CDC12 grow well with only a mild cell cycle delay. Thus, SPB- and the bud-neck-localized Cdc5p control most of the critical Cdc5p functions and downregulation of Bfa1p and Swe1p at the respective locations are two critical factors that require Cdc5p.
APA, Harvard, Vancouver, ISO, and other styles
10

Kumar, Praveen, and C. C. Wang. "Dissociation of Cytokinesis Initiation from Mitotic Control in a Eukaryote." Eukaryotic Cell 5, no. 1 (January 2006): 92–102. http://dx.doi.org/10.1128/ec.5.1.92-102.2006.

Full text
Abstract:
ABSTRACTCytokinesis is initiated only after mitotic exit in eukaryotes. However, in the insect (procyclic) form of an ancient protist,Trypanosoma brucei, a blockade at the G2/M checkpoint results in an enrichment of anucleate cells (zoids), suggesting separated regulations between mitosis and cytokinesis (X. Tu and C. C. Wang, J. Biol. Chem.279:20519-20528, 2004). Polo-like kinases (Plks) are known to play critical roles in controlling both mitosis and cytokinesis. A singlePlkhomologue inT. brucei, TbPLK, was found to be capable of complementing the Plk (Cdc5) functions inSaccharomyces cerevisiae, thus raising the question of how it may function in the trypanosome with cytokinesis dissociated from mitosis. Depletion of TbPLK in the procyclic form ofT. bruceiby RNA interference resulted in growth arrest with accumulation of multiple nuclei, kinetoplasts, basal bodies, and flagella in approximately equal numbers among individual cells. There were, however, few zoids detectable, indicating inhibited cytokinesis with unblocked mitosis and kinetoplast segregation. TbPLK is thus apparently involved only in initiating cytokinesis inT. brucei. Overexpression of TbPLK in the trypanosome did not affect cell growth, but 13% of the resulting population was in the zoid form, suggesting runaway cytokinesis. An immunofluorescence assay indicated that TbPLK was localized in a chain of likely flagellum attachment zones in the cytoskeleton. In a dividing cell, a new line of such zones appeared closely paralleling the existing one, which could constitute the cleavage furrow. An exposed region of TbPLK at the anterior tip of the cell may provide the trigger of cytokinesis. Taken together, our results revealed a novel mechanism of cytokinesis initiation in the trypanosome that may serve as a useful model for further in-depth investigations.
APA, Harvard, Vancouver, ISO, and other styles
11

Karaiskou, A., C. Jessus, T. Brassac, and R. Ozon. "Phosphatase 2A and polo kinase, two antagonistic regulators of cdc25 activation and MPF auto-amplification." Journal of Cell Science 112, no. 21 (November 1, 1999): 3747–56. http://dx.doi.org/10.1242/jcs.112.21.3747.

Full text
Abstract:
The auto-catalytic activation of the cyclin-dependent kinase Cdc2 or MPF (M-phase promoting factor) is an irreversible process responsible for the entry into M phase. In Xenopus oocyte, a positive feed-back loop between Cdc2 kinase and its activating phosphatase Cdc25 allows the abrupt activation of MPF and the entry into the first meiotic division. We have studied the Cdc2/Cdc25 feed-back loop using cell-free systems derived from Xenopus prophase-arrested oocyte. Our findings support the following two-step model for MPF amplification: during the first step, Cdc25 acquires a basal catalytic activity resulting in a linear activation of Cdc2 kinase. In turn Cdc2 partially phosphorylates Cdc25 but no amplification takes place; under this condition Plx1 kinase and its activating kinase, Plkk1 are activated. However, their activity is not required for the partial phosphorylation of Cdc25. This first step occurs independently of PP2A or Suc1/Cks-dependent Cdc25/Cdc2 association. On the contrary, the second step involves the full phosphorylation and activation of Cdc25 and the initiation of the amplification loop. It depends both on PP2A inhibition and Plx1 kinase activity. Suc1-dependent Cdc25/Cdc2 interaction is required for this process.
APA, Harvard, Vancouver, ISO, and other styles
12

Vannini, Michael, Victoria R. Mingione, Ashleigh Meyer, Courtney Sniffen, Jenna Whalen, and Anupama Seshan. "A Novel Hyperactive Nud1 Mitotic Exit Network Scaffold Causes Spindle Position Checkpoint Bypass in Budding Yeast." Cells 11, no. 1 (December 24, 2021): 46. http://dx.doi.org/10.3390/cells11010046.

Full text
Abstract:
Mitotic exit is a critical cell cycle transition that requires the careful coordination of nuclear positioning and cyclin B destruction in budding yeast for the maintenance of genome integrity. The mitotic exit network (MEN) is a Ras-like signal transduction pathway that promotes this process during anaphase. A crucial step in MEN activation occurs when the Dbf2-Mob1 protein kinase complex associates with the Nud1 scaffold protein at the yeast spindle pole bodies (SPBs; centrosome equivalents) and thereby becomes activated. This requires prior priming phosphorylation of Nud1 by Cdc15 at SPBs. Cdc15 activation, in turn, requires both the Tem1 GTPase and the Polo kinase Cdc5, but how Cdc15 associates with SPBs is not well understood. We have identified a hyperactive allele of NUD1, nud1-A308T, that recruits Cdc15 to SPBs in all stages of the cell cycle in a CDC5-independent manner. This allele leads to early recruitment of Dbf2-Mob1 during metaphase and requires known Cdc15 phospho-sites on Nud1. The presence of nud1-A308T leads to loss of coupling between nuclear position and mitotic exit in cells with mispositioned spindles. Our findings highlight the importance of scaffold regulation in signaling pathways to prevent improper activation.
APA, Harvard, Vancouver, ISO, and other styles
13

Hagan, Iain M. "The spindle pole body plays a key role in controlling mitotic commitment in the fission yeast Schizosaccharomyces pombe." Biochemical Society Transactions 36, no. 5 (September 19, 2008): 1097–101. http://dx.doi.org/10.1042/bst0361097.

Full text
Abstract:
Commitment to mitosis is regulated by a conserved protein kinase complex called MPF (mitosis-promoting factor). MPF activation triggers a positive-feedback loop that further promotes the activity of its activating phosphatase Cdc25 and is assumed to down-regulate the MPF-inhibitory kinase Wee1. Four protein kinases contribute to this amplification loop: MPF itself, Polo kinase, MAPK (mitogen-activated protein kinase) and Greatwall kinase. The fission yeast SPB (spindle pole body) component Cut12 plays a critical role in modulating mitotic commitment. In this review, I discuss the relationship between Cut12 and the fission yeast Polo kinase Plo1 in mitotic control. These results indicate that commitment to mitosis is co-ordinated by control networks on the spindle pole. I then describe how the Cut12/Plo1 control network links growth control signalling from TOR (target of rapamycin) and MAPK networks to the activation of MPF to regulate the timing of cell division.
APA, Harvard, Vancouver, ISO, and other styles
14

Mulvihill, Daniel P., Janni Petersen, Hiroyuki Ohkura, David M. Glover, and Iain M. Hagan. "Plo1 Kinase Recruitment to the Spindle Pole Body and Its Role in Cell Division inSchizosaccharomyces pombe." Molecular Biology of the Cell 10, no. 8 (August 1999): 2771–85. http://dx.doi.org/10.1091/mbc.10.8.2771.

Full text
Abstract:
Polo kinases execute multiple roles during cell division. The fission yeast polo related kinase Plo1 is required to assemble the mitotic spindle, the prophase actin ring that predicts the site for cytokinesis and for septation after the completion of mitosis ( Ohkuraet al., 1995 ; Bahler et al., 1998 ). We show that Plo1 associates with the mitotic but not interphase spindle pole body (SPB). SPB association of Plo1 is the earliest fission yeast mitotic event recorded to date. SPB association is strong from mitotic commitment to early anaphase B, after which the Plo1 signal becomes very weak and finally disappears upon spindle breakdown. SPB association of Plo1 requires mitosis-promoting factor (MPF) activity, whereas its disassociation requires the activity of the anaphase-promoting complex. The stf1.1 mutation bypasses the usual requirement for the MPF activator Cdc25 ( Hudson et al., 1990 ). Significantly, Plo1 associates inappropriately with the interphase SPB of stf1.1 cells. These data are consistent with the emerging theme from many systems that polo kinases participate in the regulation of MPF to determine the timing of commitment to mitosis and may indicate that pole association is a key aspect of Plo1 function. Plo1 does not associate with the SPB when septation is inappropriately driven by deregulation of the Spg1 pathway and remains SPB associated if septation occurs in the presence of a spindle. Thus, neither Plo1 recruitment to nor its departure from the SPB are required for septation; however, overexpression ofplo1+activates the Spg1 pathway and causes transient Cdc7 recruitment to the SPB and multiple rounds of septation.
APA, Harvard, Vancouver, ISO, and other styles
15

Cullen, C. Fiona, Karen M. May, Iain M. Hagan, David M. Glover, and Hiroyuki Ohkura. "A New Genetic Method for Isolating Functionally Interacting Genes: High plo1+-Dependent Mutants and Their Suppressors Define Genes in Mitotic and Septation Pathways in Fission Yeast." Genetics 155, no. 4 (August 1, 2000): 1521–34. http://dx.doi.org/10.1093/genetics/155.4.1521.

Full text
Abstract:
Abstract We describe a general genetic method to identify genes encoding proteins that functionally interact with and/or are good candidates for downstream targets of a particular gene product. The screen identifies mutants whose growth depends on high levels of expression of that gene. We apply this to the plo1+ gene that encodes a fission yeast homologue of the polo-like kinases. plo1+ regulates both spindle formation and septation. We have isolated 17 high plo1+-dependent (pld) mutants that show defects in mitosis or septation. Three mutants show a mitotic arrest phenotype. Among the 14 pld mutants with septation defects, 12 mapped to known loci: cdc7, cdc15, cdc11 spg1, and sid2. One of the pld mutants, cdc7-PD1, was selected for suppressor analysis. As multicopy suppressors, we isolated four known genes involved in septation in fission yeast: spg1+, sce3+, cdc8+, and rho1+, and two previously uncharacterized genes, mpd1+ and mpd2+. mpd1+ exhibits high homology to phosphatidylinositol 4-phosphate 5-kinase, while mpd2+ resembles Saccharomyces cerevisiae SMY2; both proteins are involved in the regulation of actin-mediated processes. As chromosomal suppressors of cdc7-PD1, we isolated mutations of cdc16 that resulted in multiseptation without nuclear division. cdc16+, dma1+, byr3+, byr4+ and a truncated form of the cdc7 gene were isolated by complementation of one of these cdc16 mutations. These results demonstrate that screening for high dose-dependent mutants and their suppressors is an effective approach to identify functionally interacting genes.
APA, Harvard, Vancouver, ISO, and other styles
16

Cheng, Liang, Linda Hunke, and Christopher F. J. Hardy. "Cell Cycle Regulation of the Saccharomyces cerevisiae Polo-Like Kinase Cdc5p." Molecular and Cellular Biology 18, no. 12 (December 1, 1998): 7360–70. http://dx.doi.org/10.1128/mcb.18.12.7360.

Full text
Abstract:
ABSTRACT Progression through and completion of mitosis require the actions of the evolutionarily conserved Polo kinase. We have determined that the levels of Cdc5p, a Saccharomyces cerevisiae member of the Polo family of mitotic kinases, are cell cycle regulated. Cdc5p accumulates in the nuclei of G2/M-phase cells, and its levels decline dramatically as cells progress through anaphase and begin telophase. We report that Cdc5p levels are sensitive to mutations in key components of the anaphase-promoting complex (APC). We have determined that Cdc5p-associated kinase activity is restricted to G2/M and that this activity is posttranslationally regulated. These results further link the actions of the APC to the completion of mitosis and suggest possible roles for Cdc5p during progression through and completion of mitosis.
APA, Harvard, Vancouver, ISO, and other styles
17

Hwa Lim, Hong, Foong May Yeong, and Uttam Surana. "Inactivation of Mitotic Kinase Triggers Translocation of MEN Components to Mother-Daughter Neck in Yeast." Molecular Biology of the Cell 14, no. 11 (November 2003): 4734–43. http://dx.doi.org/10.1091/mbc.e03-04-0238.

Full text
Abstract:
Chromosome segregation, mitotic exit, and cytokinesis are executed in this order during mitosis. Although a scheme coordinating sister chromatid separation and initiation of mitotic exit has been proposed, the mechanism that temporally links the onset of cytokinesis to mitotic exit is not known. Exit from mitosis is regulated by the mitotic exit network (MEN), which includes a GTPase (Tem1) and various kinases (Cdc15, Cdc5, Dbf2, and Dbf20). Here, we show that Dbf2 and Dbf20 functions are necessary for the execution of cytokinesis. Relocalization of these proteins from spindle pole bodies to mother daughter neck seems to be necessary for this role because cdc15-2 mutant cells, though capable of exiting mitosis at semipermissive temperature, are unable to localize Dbf2 (and Dbf20) to the “neck” and fail to undergo cytokinesis. These cells can assemble and constrict the actomyosin ring normally but are incapable of forming a septum, suggesting that MEN components are critical for the initiation of septum formation. Interestingly, the spindle pole body to neck translocation of Dbf2 and Dbf20 is triggered by the inactivation of mitotic kinase. The requirement of kinase inactivation for translocation of MEN components to the division site thus provides a mechanism that renders mitotic exit a prerequisite for cytokinesis.
APA, Harvard, Vancouver, ISO, and other styles
18

Botchkarev, Vladimir V., Mikael V. Garabedian, Brenda Lemos, Eric Paulissen, and James E. Haber. "The budding yeast Polo-like kinase localizes to distinct populations at centrosomes during mitosis." Molecular Biology of the Cell 28, no. 8 (April 15, 2017): 1011–20. http://dx.doi.org/10.1091/mbc.e16-05-0324.

Full text
Abstract:
The budding yeast Polo-like kinase Cdc5 is a key regulator of many mitotic events. Cdc5 coordinates its functions spatially and temporally by changing its localization during the cell cycle: Cdc5 is imported into the nucleus in G2 phase and released to the cytoplasm in anaphase, where it accumulates at the bud neck. Cdc5 also localizes to the spindle pole bodies (SPBs) from S phase until the end of mitosis. Whether Cdc5 changes its SPB population during the cell cycle is not known. We find that Cdc5 localizes to distinct SPB subpopulations, depending on the mitotic stage. Cdc5 localizes to the nuclear side of the SPBs during metaphase and early anaphase and to the cytoplasmic surface of the SPBs during late anaphase. Cdc14 is necessary to relocalize Cdc5 from the nuclear SPB plaque. Accumulation of Cdc5 at the daughter SPB in late anaphase is controlled by Bfa1. We also show that Cdc5 and Bfa1 are found in spatially distinct locations at the SPBs during G2/M arrest after DNA damage. Collectively our data reveal that Cdc5 is a dynamic component of the SPBs during mitosis and provide new insight into its regulation during both late mitotic events and DNA damage–induced G2/M arrest.
APA, Harvard, Vancouver, ISO, and other styles
19

Mortensen, Eric M., Wilhelm Haas, Melanie Gygi, Steven P. Gygi, and Douglas R. Kellogg. "Cdc28-Dependent Regulation of the Cdc5/Polo Kinase." Current Biology 15, no. 22 (November 2005): 2033–37. http://dx.doi.org/10.1016/j.cub.2005.10.046.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Zhao, Yong, Olivier Haccard, Ruoning Wang, Jiangtao Yu, Jian Kuang, Catherine Jessus, and Michael L. Goldberg. "Roles of Greatwall Kinase in the Regulation of Cdc25 Phosphatase." Molecular Biology of the Cell 19, no. 4 (April 2008): 1317–27. http://dx.doi.org/10.1091/mbc.e07-11-1099.

Full text
Abstract:
We previously reported that immunodepletion of Greatwall kinase prevents Xenopus egg extracts from entering or maintaining M phase due to the accumulation of inhibitory phosphorylations on Thr14 and Tyr15 of Cdc2. M phase–promoting factor (MPF) in turn activates Greatwall, implying that Greatwall participates in an MPF autoregulatory loop. We show here that activated Greatwall both accelerates the mitotic G2/M transition in cycling egg extracts and induces meiotic maturation in G2-arrested Xenopus oocytes in the absence of progesterone. Activated Greatwall can induce phosphorylations of Cdc25 in the absence of the activity of Cdc2, Plx1 (Xenopus Polo-like kinase) or mitogen-activated protein kinase, or in the presence of an activator of protein kinase A that normally blocks mitotic entry. The effects of active Greatwall mimic in many respects those associated with addition of the phosphatase inhibitor okadaic acid (OA); moreover, OA allows cycling extracts to enter M phase in the absence of Greatwall. Taken together, these findings support a model in which Greatwall negatively regulates a crucial phosphatase that inhibits Cdc25 activation and M phase induction.
APA, Harvard, Vancouver, ISO, and other styles
21

Song, Sukgil, and Kyung S. Lee. "A Novel Function of Saccharomyces cerevisiae CDC5 in Cytokinesis." Journal of Cell Biology 152, no. 3 (February 5, 2001): 451–70. http://dx.doi.org/10.1083/jcb.152.3.451.

Full text
Abstract:
Coordination of mitotic exit with timely initiation of cytokinesis is critical to ensure completion of mitotic events before cell division. The Saccharomyces cerevisiae polo kinase Cdc5 functions in a pathway leading to the degradation of mitotic cyclin Clb2, thereby permitting mitotic exit. Here we provide evidence that Cdc5 also plays a role in regulating cytokinesis and that an intact polo-box, a conserved motif in the noncatalytic COOH-terminal domain of Cdc5, is required for this event. Depletion of Cdc5 function leads to an arrest in cytokinesis. Overexpression of the COOH-terminal domain of Cdc5 (cdc5ΔN), but not the corresponding polo-box mutant, resulted in connected cells. These cells shared cytoplasms with incomplete septa, and possessed aberrant septin ring structures. Provision of additional copies of endogenous CDC5 remedied this phenotype, suggesting a dominant-negative inhibition of cytokinesis. The polo-box–dependent interactions between Cdc5 and septins (Cdc11 and Cdc12) and genetic interactions between the dominant-negative cdc5ΔN and Cyk2/Hof1 or Myo1 suggest that direct interactions between cdc5ΔN and septins resulted in inhibition of Cyk2/Hof1- and Myo1-mediated cytokinetic pathways. Thus, we propose that Cdc5 may coordinate mitotic exit with cytokinesis by participating in both anaphase promoting complex activation and a polo-box–dependent cytokinetic pathway.
APA, Harvard, Vancouver, ISO, and other styles
22

Luca, Francis C., and Mark Winey. "MOB1, an Essential Yeast Gene Required for Completion of Mitosis and Maintenance of Ploidy." Molecular Biology of the Cell 9, no. 1 (January 1998): 29–46. http://dx.doi.org/10.1091/mbc.9.1.29.

Full text
Abstract:
Mob1p is an essential Saccharomyces cerevisiaeprotein, identified from a two-hybrid screen, that binds Mps1p, a protein kinase essential for spindle pole body duplication and mitotic checkpoint regulation. Mob1p contains no known structural motifs; however MOB1 is a member of a conserved gene family and shares sequence similarity with a nonessential yeast gene,MOB2. Mob1p is a phosphoprotein in vivo and a substrate for the Mps1p kinase in vitro. Conditional alleles ofMOB1 cause a late nuclear division arrest at restrictive temperature. MOB1 exhibits genetic interaction with three other yeast genes required for the completion of mitosis,LTE1, CDC5, and CDC15 (the latter two encode essential protein kinases). Most haploid mutantmob1 strains also display a complete increase in ploidy at permissive temperature. The mechanism for the increase in ploidy may occur through MPS1 function. One mob1strain, which maintains stable haploidy at both permissive and restrictive temperature, diploidizes at permissive temperature when combined with the mps1–1 mutation. Strains containingmob2Δ also display a complete increase in ploidy when combined with the mps1-1 mutation. Perhaps in addition to, or as part of, its essential function in late mitosis, MOB1 is required for a cell cycle reset function necessary for the initiation of the spindle pole body duplication.
APA, Harvard, Vancouver, ISO, and other styles
23

Masuda, Hirohisa, Chii Shyang Fong, Chizuru Ohtsuki, Tokuko Haraguchi, and Yasushi Hiraoka. "Spatiotemporal regulations of Wee1 at the G2/M transition." Molecular Biology of the Cell 22, no. 5 (March 2011): 555–69. http://dx.doi.org/10.1091/mbc.e10-07-0644.

Full text
Abstract:
Wee1 is a protein kinase that negatively regulates mitotic entry in G2 phase by suppressing cyclin B–Cdc2 activity, but its spatiotemporal regulations remain to be elucidated. We observe the dynamic behavior of Wee1 in Schizosaccharomyces pombe cells and manipulate its localization and kinase activity to study its function. At late G2, nuclear Wee1 efficiently suppresses cyclin B–Cdc2 around the spindle pole body (SPB). During the G2/M transition when cyclin B–Cdc2 is highly enriched at the SPB, Wee1 temporally accumulates at the nuclear face of the SPB in a cyclin B–Cdc2-dependent manner and locally suppresses both cyclin B–Cdc2 activity and spindle assembly to counteract a Polo kinase–dependent positive feedback loop. Then Wee1 disappears from the SPB during spindle assembly. We propose that regulation of Wee1 localization around the SPB during the G2/M transition is important for proper mitotic entry and progression.
APA, Harvard, Vancouver, ISO, and other styles
24

Maekawa, Hiromi, Shen Jiangyan, Kaoru Takegawa, and Gislene Pereira. "SIN-like Pathway Kinases Regulate the End of Mitosis in the Methylotrophic Yeast Ogataea polymorpha." Cells 11, no. 9 (April 30, 2022): 1519. http://dx.doi.org/10.3390/cells11091519.

Full text
Abstract:
The mitotic exit network (MEN) is a conserved signalling pathway essential for the termination of mitosis in the budding yeast Saccharomyces cerevisiae. All MEN components are highly conserved in the methylotrophic budding yeast Ogataea polymorpha, except for Cdc15 kinase. Instead, we identified two essential kinases OpHcd1 and OpHcd2 (homologue candidate of ScCdc15) that are homologous to SpSid1 and SpCdc7, respectively, components of the septation initiation network (SIN) of the fission yeast Schizosaccharomyces pombe. Conditional mutants for OpHCD1 and OpHCD2 exhibited significant delay in late anaphase and defective cell separation, suggesting that both genes have roles in mitotic exit and cytokinesis. Unlike Cdc15 in S. cerevisiae, the association of OpHcd1 and OpHcd2 with the yeast centrosomes (named spindle pole bodies, SPBs) is restricted to the SPB in the mother cell body. SPB localisation of OpHcd2 is regulated by the status of OpTem1 GTPase, while OpHcd1 requires the polo-like kinase OpCdc5 as well as active Tem1 to ensure the coordination of mitotic exit (ME) signalling and cell cycle progression. Our study suggests that the divergence of molecular mechanisms to control the ME-signalling pathway as well as the loss of Sid1/Hcd1 kinase in the MEN occurred relatively recently during the evolution of budding yeast.
APA, Harvard, Vancouver, ISO, and other styles
25

Khondker, Shoily, Sam Kajjo, Devon Chandler-Brown, Jan Skotheim, Adam Rudner, and Amy Ikui. "PP2ACdc55 dephosphorylates Pds1 and inhibits spindle elongation in S. cerevisiae." Journal of Cell Science 133, no. 14 (June 26, 2020): jcs243766. http://dx.doi.org/10.1242/jcs.243766.

Full text
Abstract:
ABSTRACTPP2ACdc55 (the form of protein phosphatase 2A containing Cdc55) regulates cell cycle progression by reversing cyclin-dependent kinase (CDK)- and polo-like kinase (Cdc5)-dependent phosphorylation events. In S. cerevisiae, Cdk1 phosphorylates securin (Pds1), which facilitates Pds1 binding and inhibits separase (Esp1). During anaphase, Esp1 cleaves the cohesin subunit Scc1 and promotes spindle elongation. Here, we show that PP2ACdc55 directly dephosphorylates Pds1 both in vivo and in vitro. Pds1 hyperphosphorylation in a cdc55 deletion mutant enhanced the Pds1–Esp1 interaction, which played a positive role in Pds1 nuclear accumulation and in spindle elongation. We also show that nuclear PP2ACdc55 plays a role during replication stress to inhibit spindle elongation. This pathway acted independently of the known Mec1, Swe1 or spindle assembly checkpoint (SAC) checkpoint pathways. We propose a model where Pds1 dephosphorylation by PP2ACdc55 disrupts the Pds1–Esp1 protein interaction and inhibits Pds1 nuclear accumulation, which prevents spindle elongation, a process that is elevated during replication stress.
APA, Harvard, Vancouver, ISO, and other styles
26

Acosta, Isabel, David Ontoso, and Pedro A. San-Segundo. "The budding yeast polo-like kinase Cdc5 regulates the Ndt80 branch of the meiotic recombination checkpoint pathway." Molecular Biology of the Cell 22, no. 18 (September 15, 2011): 3478–90. http://dx.doi.org/10.1091/mbc.e11-06-0482.

Full text
Abstract:
Defects in chromosome synapsis and/or meiotic recombination activate a surveillance mechanism that blocks meiotic cell cycle progression to prevent anomalous chromosome segregation and formation of aberrant gametes. In the budding yeast zip1 mutant, which lacks a synaptonemal complex component, the meiotic recombination checkpoint is triggered, resulting in extremely delayed meiotic progression. We report that overproduction of the polo-like kinase Cdc5 partially alleviates the meiotic prophase arrest of zip1, leading to the formation of inviable meiotic products. Unlike vegetative cells, we demonstrate that Cdc5 overproduction does not stimulate meiotic checkpoint adaptation because the Mek1 kinase remains activated in zip1 2μ-CDC5 cells. Inappropriate meiotic divisions in zip1 promoted by high levels of active Cdc5 do not result from altered function of the cyclin-dependent kinase (CDK) inhibitor Swe1. In contrast, CDC5 overexpression leads to premature induction of the Ndt80 transcription factor, which drives the expression of genes required for meiotic divisions, including CLB1. We also show that depletion of Cdc5 during meiotic prophase prevents the production of Ndt80 and that CDK activity contributes to the induction of Ndt80 in zip1 cells overexpressing CDC5. Our results reveal a role for Cdc5 in meiotic checkpoint control by regulating Ndt80 function.
APA, Harvard, Vancouver, ISO, and other styles
27

Liang, Fengshan, Fengzhi Jin, Hong Liu, and Yanchang Wang. "The Molecular Function of the Yeast Polo-like Kinase Cdc5 in Cdc14 Release during Early Anaphase." Molecular Biology of the Cell 20, no. 16 (August 15, 2009): 3671–79. http://dx.doi.org/10.1091/mbc.e08-10-1049.

Full text
Abstract:
In the budding yeast Saccharomyces cerevisiae , Cdc14 is sequestered within the nucleolus before anaphase entry through its association with Net1/Cfi1, a nucleolar protein. Protein phosphatase PP2ACdc55 dephosphorylates Net1 and keeps it as a hypophosphorylated form before anaphase. Activation of the Cdc fourteen early anaphase release (FEAR) pathway after anaphase entry induces a brief Cdc14 release from the nucleolus. Some of the components in the FEAR pathway, including Esp1, Slk19, and Spo12, inactivate PP2ACdc55, allowing the phosphorylation of Net1 by mitotic cyclin-dependent kinase (Cdk) (Clb2-Cdk1). However, the function of another FEAR component, the Polo-like kinase Cdc5, remains elusive. Here, we show evidence indicating that Cdc5 promotes Cdc14 release primarily by stimulating the degradation of Swe1, the inhibitory kinase for mitotic Cdk. First, we found that deletion of SWE1 partially suppresses the FEAR defects in cdc5 mutants. In contrast, high levels of Swe1 impair FEAR activation. We also demonstrated that the accumulation of Swe1 in cdc5 mutants is responsible for the decreased Net1 phosphorylation. Therefore, we conclude that the down-regulation of Swe1 protein levels by Cdc5 promotes FEAR activation by relieving the inhibition on Clb2-Cdk1, the kinase for Net1 protein.
APA, Harvard, Vancouver, ISO, and other styles
28

Liang, Jun, and Peter Fantes. "The Schizosaccharomyces pombe Cdc7 Protein Kinase Required for Septum Formation Is a Client Protein of Cdc37." Eukaryotic Cell 6, no. 7 (May 11, 2007): 1089–96. http://dx.doi.org/10.1128/ec.00080-07.

Full text
Abstract:
ABSTRACT Cdc37 is an essential molecular chaperone found in fungi and metazoa whose main specificity is for certain protein kinases. Cdc37 can act as an Hsp90 cochaperone or alone; in yeasts, the interaction with Hsp90 is weak and appears not to be essential for Cdc37 function. Numerous genetic interactions between Cdc37 and likely client proteins have been observed in yeasts, but biochemical confirmation has been reported in only a few cases. We and others have generated and characterized temperature-sensitive cdc37 alleles in S. pombe and have used them to investigate the cellular roles of Cdc37: previous work has shown that mitotic Cdc2 is a major client. In this paper, we describe a screen for mutations synthetically lethal with a cdc37ts mutant with the aim of identifying genes encoding further client proteins of Cdc37. Ten such strains were isolated, and genomic libraries were screened for rescuing plasmids. In one case, a truncated cdc7 gene was identified. Further experiments showed that the mutation in this strain was indeed in cdc7. Cdc7 is a protein kinase required for septum initiation, and we show that its kinase activity is greatly reduced when Cdc37 function is impaired. Cdc7 normally locates to the spindle pole body during mitosis, and this appears to be unaffected in the cdc37ts mutant. Other evidence suggests that, in addition to mitosis and septum initiation, Cdc37 may also be required for septum cleavage.
APA, Harvard, Vancouver, ISO, and other styles
29

Mishra, Prashant K., Sultan Ciftci-Yilmaz, David Reynolds, Wei-Chun Au, Lars Boeckmann, Lauren E. Dittman, Ziad Jowhar, et al. "Polo kinase Cdc5 associates with centromeres to facilitate the removal of centromeric cohesin during mitosis." Molecular Biology of the Cell 27, no. 14 (July 15, 2016): 2286–300. http://dx.doi.org/10.1091/mbc.e16-01-0004.

Full text
Abstract:
Sister chromatid cohesion is essential for tension-sensing mechanisms that monitor bipolar attachment of replicated chromatids in metaphase. Cohesion is mediated by the association of cohesins along the length of sister chromatid arms. In contrast, centromeric cohesin generates intrastrand cohesion and sister centromeres, while highly cohesin enriched, are separated by >800 nm at metaphase in yeast. Removal of cohesin is necessary for sister chromatid separation during anaphase, and this is regulated by evolutionarily conserved polo-like kinase (Cdc5 in yeast, Plk1 in humans). Here we address how high levels of cohesins at centromeric chromatin are removed. Cdc5 associates with centromeric chromatin and cohesin-associated regions. Maximum enrichment of Cdc5 in centromeric chromatin occurs during the metaphase-to-anaphase transition and coincides with the removal of chromosome-associated cohesin. Cdc5 interacts with cohesin in vivo, and cohesin is required for association of Cdc5 at centromeric chromatin. Cohesin removal from centromeric chromatin requires Cdc5 but removal at distal chromosomal arm sites does not. Our results define a novel role for Cdc5 in regulating removal of centromeric cohesins and faithful chromosome segregation.
APA, Harvard, Vancouver, ISO, and other styles
30

George, Vinoj T., Gavin Brooks, and Timothy C. Humphrey. "Regulation of Cell Cycle and Stress Responses to Hydrostatic Pressure in Fission Yeast." Molecular Biology of the Cell 18, no. 10 (October 2007): 4168–79. http://dx.doi.org/10.1091/mbc.e06-12-1141.

Full text
Abstract:
We have investigated the cellular responses to hydrostatic pressure by using the fission yeast Schizosaccharomyces pombe as a model system. Exposure to sublethal levels of hydrostatic pressure resulted in G2 cell cycle delay. This delay resulted from Cdc2 tyrosine-15 (Y-15) phosphorylation, and it was abrogated by simultaneous disruption of the Cdc2 kinase regulators Cdc25 and Wee1. However, cell cycle delay was independent of the DNA damage, cytokinesis, and cell size checkpoints, suggesting a novel mechanism of Cdc2-Y15 phosphorylation in response to hydrostatic pressure. Spc1/Sty1 mitogen-activated protein (MAP) kinase, a conserved member of the eukaryotic stress-activated p38, mitogen-activated protein (MAP) kinase family, was rapidly activated after pressure stress, and it was required for cell cycle recovery under these conditions, in part through promoting polo kinase (Plo1) phosphorylation on serine 402. Moreover, the Spc1 MAP kinase pathway played a key role in maintaining cell viability under hydrostatic pressure stress through the bZip transcription factor, Atf1. Further analysis revealed that prestressing cells with heat increased barotolerance, suggesting adaptational cross-talk between these stress responses. These findings provide new insight into eukaryotic homeostasis after exposure to pressure stress.
APA, Harvard, Vancouver, ISO, and other styles
31

González-Arranz, Sara, Isabel Acosta, Jesús A. Carballo, Beatriz Santos, and Pedro A. San-Segundo. "The N-Terminal Region of the Polo Kinase Cdc5 Is Required for Downregulation of the Meiotic Recombination Checkpoint." Cells 10, no. 10 (September 27, 2021): 2561. http://dx.doi.org/10.3390/cells10102561.

Full text
Abstract:
During meiosis, the budding yeast polo-like kinase Cdc5 is a crucial driver of the prophase I to meiosis I (G2/M) transition. The meiotic recombination checkpoint restrains cell cycle progression in response to defective recombination to ensure proper distribution of intact chromosomes to the gametes. This checkpoint detects unrepaired DSBs and initiates a signaling cascade that ultimately inhibits Ndt80, a transcription factor required for CDC5 gene expression. Previous work revealed that overexpression of CDC5 partially alleviates the checkpoint-imposed meiotic delay in the synaptonemal complex-defective zip1Δ mutant. Here, we show that overproduction of a Cdc5 version (Cdc5-ΔN70), lacking the N-terminal region required for targeted degradation of the protein by the APC/C complex, fails to relieve the zip1Δ-induced meiotic delay, despite being more stable and reaching increased protein levels. However, precise mutation of the consensus motifs for APC/C recognition (D-boxes and KEN) has no effect on Cdc5 stability or function during meiosis. Compared to the zip1Δ single mutant, the zip1Δ cdc5-ΔN70 double mutant exhibits an exacerbated meiotic block and reduced levels of Ndt80 consistent with persistent checkpoint activity. Finally, using a CDC5-inducible system, we demonstrate that the N-terminal region of Cdc5 is essential for its checkpoint erasing function. Thus, our results unveil an additional layer of regulation of polo-like kinase function in meiotic cell cycle control.
APA, Harvard, Vancouver, ISO, and other styles
32

Pakchuen, Sujiraporn, Mai Ishibashi, Emi Takakusagi, Katsuhiko Shirahige, and Takashi Sutani. "Physical Association of Saccharomyces cerevisiae Polo-like Kinase Cdc5 with Chromosomal Cohesin Facilitates DNA Damage Response." Journal of Biological Chemistry 291, no. 33 (June 20, 2016): 17228–46. http://dx.doi.org/10.1074/jbc.m116.727438.

Full text
Abstract:
At the onset of anaphase, a protease called separase breaks the link between sister chromatids by cleaving the cohesin subunit Scc1. This irreversible step in the cell cycle is promoted by degradation of the separase inhibitor, securin, and polo-like kinase (Plk) 1-dependent phosphorylation of the Scc1 subunit. Plk could recognize substrates through interaction between its phosphopeptide interaction domain, the polo-box domain, and a phosphorylated priming site in the substrate, which has been generated by a priming kinase beforehand. However, the physiological relevance of this targeting mechanism remains to be addressed for many of the Plk1 substrates. Here, we show that budding yeast Plk1, Cdc5, is pre-deposited onto cohesin engaged in cohesion on chromosome arms in G2/M phase cells. The Cdc5-cohesin association is mediated by direct interaction between the polo-box domain of Cdc5 and Scc1 phosphorylated at multiple sites in its middle region. Alanine substitutions of the possible priming phosphorylation sites (scc1-15A) impair Cdc5 association with chromosomal cohesin, but they make only a moderate impact on mitotic cell growth even in securin-deleted cells (pds1Δ), where Scc1 phosphorylation by Cdc5 is indispensable. The same scc1-15A pds1Δ double mutant, however, exhibits marked sensitivity to the DNA-damaging agent phleomycin, suggesting that the priming phosphorylation of Scc1 poses an additional layer of regulation that enables yeast cells to adapt to genotoxic environments.
APA, Harvard, Vancouver, ISO, and other styles
33

Attner, M. A., M. P. Miller, L. s. Ee, S. K. Elkin, and A. Amon. "Polo kinase Cdc5 is a central regulator of meiosis I." Proceedings of the National Academy of Sciences 110, no. 35 (August 5, 2013): 14278–83. http://dx.doi.org/10.1073/pnas.1311845110.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Mishra, Prashant K., Gudjon Olafsson, Lars Boeckmann, Timothy J. Westlake, Ziad M. Jowhar, Lauren E. Dittman, Richard E. Baker, Damien D’Amours, Peter H. Thorpe, and Munira A. Basrai. "Cell cycle–dependent association of polo kinase Cdc5 with CENP-A contributes to faithful chromosome segregation in budding yeast." Molecular Biology of the Cell 30, no. 8 (April 2019): 1020–36. http://dx.doi.org/10.1091/mbc.e18-09-0584.

Full text
Abstract:
Evolutionarily conserved polo-like kinase, Cdc5 (Plk1 in humans), associates with kinetochores during mitosis; however, the role of cell cycle–dependent centromeric ( CEN) association of Cdc5 and its substrates that exclusively localize to the kinetochore have not been characterized. Here we report that evolutionarily conserved CEN histone H3 variant, Cse4 (CENP-A in humans), is a substrate of Cdc5, and that the cell cycle–regulated association of Cse4 with Cdc5 is required for cell growth. Cdc5 contributes to Cse4 phosphorylation in vivo and interacts with Cse4 in mitotic cells. Mass spectrometry analysis of in vitro kinase assays showed that Cdc5 phosphorylates nine serine residues clustered within the N-terminus of Cse4. Strains with cse4-9SA exhibit increased errors in chromosome segregation, reduced levels of CEN-associated Mif2 and Mcd1/Scc1 when combined with a deletion of MCM21. Moreover, the loss of Cdc5 from the CEN chromatin contributes to defects in kinetochore integrity and reduction in CEN-associated Cse4. The cell cycle–regulated association of Cdc5 with Cse4 is essential for cell viability as constitutive association of Cdc5 with Cse4 at the kinetochore leads to growth defects. In summary, our results have defined a role for Cdc5-mediated Cse4 phosphorylation in faithful chromosome segregation.
APA, Harvard, Vancouver, ISO, and other styles
35

Visintin, Rosella, Frank Stegmeier, and Angelika Amon. "The Role of the Polo Kinase Cdc5 in Controlling Cdc14 Localization." Molecular Biology of the Cell 14, no. 11 (November 2003): 4486–98. http://dx.doi.org/10.1091/mbc.e03-02-0095.

Full text
Abstract:
In budding yeast, the protein phosphatase Cdc14 controls exit from mitosis. Its activity is regulated by a competitive inhibitor Cfi1/Net1, which binds to and sequesters Cdc14 in the nucleolus. During anaphase, Cdc14 is released from its inhibitor by the action of two regulatory networks. The Cdc Fourteen Early Anaphase Release (FEAR) network initiates Cdc14 release from Cfi1/Net1 during early anaphase, and the Mitotic Exit Network (MEN) promotes Cdc14 release during late anaphase. Here, we investigate the relationship among FEAR network components and propose an order in which they function to promote Cdc14 release from the nucleolus. Furthermore, we examine the role of the protein kinase Cdc5, which is a component of both the FEAR network and the MEN, in Cdc14 release from the nucleolus. We find that overexpression of CDC5 led to Cdc14 release from the nucleolus in S phase-arrested cells, which correlated with the appearance of phosphorylated forms of Cdc14 and Cfi1/Net1. Cdc5 promotes Cdc14 phosphorylation and, by stimulating the MEN, Cfi1/Net1 phosphorylation. Furthermore, we suggest that Cdc14 release from the nucleolus only occurs when Cdc14 and Cfi1/Net1 are both phosphorylated.
APA, Harvard, Vancouver, ISO, and other styles
36

Golsteyn, R. M., K. E. Mundt, A. M. Fry, and E. A. Nigg. "Cell cycle regulation of the activity and subcellular localization of Plk1, a human protein kinase implicated in mitotic spindle function." Journal of Cell Biology 129, no. 6 (June 15, 1995): 1617–28. http://dx.doi.org/10.1083/jcb.129.6.1617.

Full text
Abstract:
Correct assembly and function of the mitotic spindle during cell division is essential for the accurate partitioning of the duplicated genome to daughter cells. Protein phosphorylation has long been implicated in controlling spindle function and chromosome segregation, and genetic studies have identified several protein kinases and phosphatases that are likely to regulate these processes. In particular, mutations in the serine/threonine-specific Drosophila kinase polo, and the structurally related kinase Cdc5p of Saccharomyces cerevisae, result in abnormal mitotic and meiotic divisions. Here, we describe a detailed analysis of the cell cycle-dependent activity and subcellular localization of Plk1, a recently identified human protein kinase with extensive sequence similarity to both Drosophila polo and S. cerevisiae Cdc5p. With the aid of recombinant baculoviruses, we have established a reliable in vitro assay for Plk1 kinase activity. We show that the activity of human Plk1 is cell cycle regulated, Plk1 activity being low during interphase but high during mitosis. We further show, by immunofluorescent confocal laser scanning microscopy, that human Plk1 binds to components of the mitotic spindle at all stages of mitosis, but undergoes a striking redistribution as cells progress from metaphase to anaphase. Specifically, Plk1 associates with spindle poles up to metaphase, but relocalizes to the equatorial plane, where spindle microtubules overlap (the midzone), as cells go through anaphase. These results indicate that the association of Plk1 with the spindle is highly dynamic and that Plk1 may function at multiple stages of mitotic progression. Taken together, our data strengthen the notion that human Plk1 may represent a functional homolog of polo and Cdc5p, and they suggest that this kinase plays an important role in the dynamic function of the mitotic spindle during chromosome segregation.
APA, Harvard, Vancouver, ISO, and other styles
37

Visintin, Rosella, and Angelika Amon. "Regulation of the Mitotic Exit Protein Kinases Cdc15 and Dbf2." Molecular Biology of the Cell 12, no. 10 (October 2001): 2961–74. http://dx.doi.org/10.1091/mbc.12.10.2961.

Full text
Abstract:
In budding yeast, the release of the protein phosphatase Cdc14 from its inhibitor Cfi1/Net1 in the nucleolus during anaphase triggers the inactivation of Clb CDKs that leads to exit from mitosis. The mitotic exit pathway controls the association between Cdc14 and Cfi1/Net1. It is comprised of the RAS-like GTP binding protein Tem1, the exchange factor Lte1, the GTPase activating protein complex Bub2-Bfa1/Byr4, and several protein kinases including Cdc15 and Dbf2. Here we investigate the regulation of the protein kinases Dbf2 and Cdc15. We find that Cdc15 is recruited to both spindle pole bodies (SPBs) during anaphase. This recruitment depends on TEM1 but notDBF2 or CDC14 and is inhibited byBUB2. Dbf2 also localizes to SPBs during anaphase, which coincides with activation of Dbf2 kinase activity. Both events depend on the mitotic exit pathway components TEM1 andCDC15. In cells lacking BUB2, Dbf2 localized to SPBs in cell cycle stages other than anaphase and telophase and Dbf2 kinase was prematurely active during metaphase. Our results suggest an order of function of mitotic exit pathway components with respect to SPB localization of Cdc15 and Dbf2 and activation of Dbf2 kinase. BUB2 negatively regulates all 3 events. Loading of Cdc15 on SPBs depends on TEM1, whereas loading of Dbf2 on SPBs and activation of Dbf2 kinase depend onTEM1 and CDC15.
APA, Harvard, Vancouver, ISO, and other styles
38

Cai, Ti, Jason Aulds, Tina Gill, Michael Cerio, and Mark E. Schmitt. "The Saccharomyces cerevisiae RNase Mitochondrial RNA Processing Is Critical for Cell Cycle Progression at the End of Mitosis." Genetics 161, no. 3 (July 1, 2002): 1029–42. http://dx.doi.org/10.1093/genetics/161.3.1029.

Full text
Abstract:
Abstract We have identified a cell cycle delay in Saccharomyces cerevisiae RNase MRP mutants. Mutants delay with large budded cells, dumbbell-shaped nuclei, and extended spindles characteristic of “exit from mitosis” mutants. In accord with this, a RNase MRP mutation can be suppressed by overexpressing the polo-like kinase CDC5 or by deleting the B-type cyclin CLB1, without restoring the MRP-dependent rRNA-processing step. In addition, we identified a series of genetic interactions between RNase MRP mutations and mutations in CDC5, CDC14, CDC15, CLB2, and CLB5. As in most “exit from mitosis” mutants, levels of the Clb2 cyclin were increased. The buildup of Clb2 protein is not the result of a defect in the release of the Cdc14 phosphatase from the nucleolus, but rather the result of an increase in CLB2 mRNA levels. These results indicate a clear role of RNase MRP in cell cycle progression at the end of mitosis. Conservation of this function in humans may explain many of the pleiotropic phenotypes of cartilage hair hypoplasia.
APA, Harvard, Vancouver, ISO, and other styles
39

Iacovella, Maria G., Catherine N. Daly, Jamie-Sioux Kelly, Adriana J. Michielsen, and Rosemary K. Clyne. "Analysis of Polo-like kinase Cdc5 in the meiosis recombination checkpoint." Cell Cycle 9, no. 6 (March 15, 2010): 1182–93. http://dx.doi.org/10.4161/cc.9.6.11068.

Full text
APA, Harvard, Vancouver, ISO, and other styles
40

Wang, S., B. Xu, L. C. Liou, Q. Ren, S. Huang, Y. Luo, Z. Zhang, and S. N. Witt. "-Synuclein disrupts stress signaling by inhibiting polo-like kinase Cdc5/Plk2." Proceedings of the National Academy of Sciences 109, no. 40 (September 17, 2012): 16119–24. http://dx.doi.org/10.1073/pnas.1206286109.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

Lee, K. S., Y. L. Yuan, R. Kuriyama, and R. L. Erikson. "Plk is an M-phase-specific protein kinase and interacts with a kinesin-like protein, CHO1/MKLP-1." Molecular and Cellular Biology 15, no. 12 (December 1995): 7143–51. http://dx.doi.org/10.1128/mcb.15.12.7143.

Full text
Abstract:
PLK (STPK13) encodes a murine protein kinase closely related to those encoded by the Drosophila melanogaster polo gene and the Saccharomyces cerevisiae CDC5 gene, which are required for normal mitotic and meiotic divisions. Affinity-purified antibody generated against the C-terminal 13 amino acids of Plk specifically recognizes a single polypeptide of 66 kDa in MELC, NIH 3T3, and HeLa cellular extracts. The expression levels of both poly(A)+ PLK mRNA and its encoded protein are most abundant about 17 h after serum stimulation of NIH 3T3 cells. Plk protein begins to accumulate at the S/G2 boundary and reaches the maximum level at the G2/M boundary in continuously cycling cells. Concurrent with cyclin B-associated cdc2 kinase activity, Plk kinase activity sharply peaks at the onset of mitosis. Plk enzymatic activity gradually decreases as M phase proceeds but persists longer than cyclin B-associated cdc2 kinase activity. Plk is localized to the area surrounding the chromosomes in prometaphase, appears condensed as several discrete bands along the spindle axis at the interzone in anaphase, and finally concentrates at the midbody during telophase and cytokinesis. Plk and CHO1/mitotic kinesin-like protein 1 (MKLP-1), which induces microtubule bundling and antiparallel movement in vitro, are colocalized during late M phase. In addition, CHO1/MKLP-1 appears to interact with Plk in vivo and to be phosphorylated by Plk-associated kinase activity in vitro.
APA, Harvard, Vancouver, ISO, and other styles
42

Luca, Francis C., Manali Mody, Cornelia Kurischko, David M. Roof, Thomas H. Giddings, and Mark Winey. "Saccharomyces cerevisiae Mob1p Is Required for Cytokinesis and Mitotic Exit." Molecular and Cellular Biology 21, no. 20 (October 15, 2001): 6972–83. http://dx.doi.org/10.1128/mcb.21.20.6972-6983.2001.

Full text
Abstract:
ABSTRACT The Saccharomyces cerevisiae mitotic exit network (MEN) is a conserved set of genes that mediate the transition from mitosis to G1 by regulating mitotic cyclin degradation and the inactivation of cyclin-dependent kinase (CDK). Here, we demonstrate that, in addition to mitotic exit, S. cerevisiae MEN gene MOB1 is required for cytokinesis and cell separation. The cytokinesis defect was evident in mob1mutants under conditions in which there was no mitotic-exit defect. Observation of live cells showed that yeast myosin II, Myo1p, was present in the contractile ring at the bud neck but that the ring failed to contract and disassemble. The cytokinesis defect persisted for several mitotic cycles, resulting in chains of cells with correctly segregated nuclei but with uncontracted actomyosin rings. The cytokinesis proteins Cdc3p (a septin), actin, and Iqg1p/ Cyk1p (an IQGAP-like protein) appeared to correctly localize inmob1 mutants, suggesting that MOB1functions subsequent to actomyosin ring assembly. We also examined the subcellular distribution of Mob1p during the cell cycle and found that Mob1p first localized to the spindle pole bodies during mid-anaphase and then localized to a ring at the bud neck just before and during cytokinesis. Localization of Mob1p to the bud neck requiredCDC3, MEN genes CDC5,CDC14, CDC15, and DBF2, and spindle pole body gene NUD1 but was independent ofMYO1. The localization of Mob1p to both spindle poles was abolished in cdc15 and nud1 mutants and was perturbed in cdc5 and cdc14mutants. These results suggest that the MEN functions during the mitosis-to-G1 transition to control cyclin-CDK inactivation and cytokinesis.
APA, Harvard, Vancouver, ISO, and other styles
43

Furstenthal, Laura, Brett K. Kaiser, Craig Swanson, and Peter K. Jackson. "Cyclin E Uses Cdc6 as a Chromatin-Associated Receptor Required for DNA Replication." Journal of Cell Biology 152, no. 6 (March 19, 2001): 1267–78. http://dx.doi.org/10.1083/jcb.152.6.1267.

Full text
Abstract:
Using an in vitro chromatin assembly assay in Xenopus egg extract, we show that cyclin E binds specifically and saturably to chromatin in three phases. In the first phase, the origin recognition complex and Cdc6 prereplication proteins, but not the minichromosome maintenance complex, are necessary and biochemically sufficient for ATP-dependent binding of cyclin E–Cdk2 to DNA. We find that cyclin E binds the NH2-terminal region of Cdc6 containing Cy–Arg-X-Leu (RXL) motifs. Cyclin E proteins with mutated substrate selection (Met-Arg-Ala-Ile-Leu; MRAIL) motifs fail to bind Cdc6, fail to compete with endogenous cyclin E–Cdk2 for chromatin binding, and fail to rescue replication in cyclin E–depleted extracts. Cdc6 proteins with mutations in the three consensus RXL motifs are quantitatively deficient for cyclin E binding and for rescuing replication in Cdc6-depleted extracts. Thus, the cyclin E–Cdc6 interaction that localizes the Cdk2 complex to chromatin is important for DNA replication. During the second phase, cyclin E–Cdk2 accumulates on chromatin, dependent on polymerase activity. In the third phase, cyclin E is phosphorylated, and the cyclin E–Cdk2 complex is displaced from chromatin in mitosis. In vitro, mitogen-activated protein kinase and especially cyclin B–Cdc2, but not the polo-like kinase 1, remove cyclin E–Cdk2 from chromatin. Rebinding of hyperphosphorylated cyclin E–Cdk2 to interphase chromatin requires dephosphorylation, and the Cdk kinase–directed Cdc14 phosphatase is sufficient for this dephosphorylation in vitro. These three phases of cyclin E association with chromatin may facilitate the diverse activities of cyclin E–Cdk2 in initiating replication, blocking rereplication, and allowing resetting of origins after mitosis.
APA, Harvard, Vancouver, ISO, and other styles
44

Sourirajan, A., and M. Lichten. "Polo-like kinase Cdc5 drives exit from pachytene during budding yeast meiosis." Genes & Development 22, no. 19 (October 1, 2008): 2627–32. http://dx.doi.org/10.1101/gad.1711408.

Full text
APA, Harvard, Vancouver, ISO, and other styles
45

Rahal, Rami, and Angelika Amon. "The Polo-like kinase Cdc5 interacts with FEAR network components and Cdc14." Cell Cycle 7, no. 20 (October 15, 2008): 3262–72. http://dx.doi.org/10.4161/cc.7.20.6852.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Lee, B. H. "Role of Polo-like Kinase CDC5 in Programming Meiosis I Chromosome Segregation." Science 300, no. 5618 (March 27, 2003): 482–86. http://dx.doi.org/10.1126/science.1081846.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Walters, Alison D., Christopher K. May, Emma S. Dauster, Bertrand P. Cinquin, Elizabeth A. Smith, Xavier Robellet, Damien D’Amours, Carolyn A. Larabell, and Orna Cohen-Fix. "The Yeast Polo Kinase Cdc5 Regulates the Shape of the Mitotic Nucleus." Current Biology 24, no. 23 (December 2014): 2861–67. http://dx.doi.org/10.1016/j.cub.2014.10.029.

Full text
APA, Harvard, Vancouver, ISO, and other styles
48

Maekawa, Hiromi, Claire Priest, Johannes Lechner, Gislene Pereira, and Elmar Schiebel. "The yeast centrosome translates the positional information of the anaphase spindle into a cell cycle signal." Journal of Cell Biology 179, no. 3 (October 29, 2007): 423–36. http://dx.doi.org/10.1083/jcb.200705197.

Full text
Abstract:
The spindle orientation checkpoint (SPOC) of budding yeast delays mitotic exit when cytoplasmic microtubules (MTs) are defective, causing the spindle to become misaligned. Delay is achieved by maintaining the activity of the Bfa1–Bub2 guanosine triphosphatase–activating protein complex, an inhibitor of mitotic exit. In this study, we show that the spindle pole body (SPB) component Spc72, a transforming acidic coiled coil–like molecule that interacts with the γ-tubulin complex, recruits Kin4 kinase to both SPBs when cytoplasmic MTs are defective. This allows Kin4 to phosphorylate the SPB-associated Bfa1, rendering it resistant to inactivation by Cdc5 polo kinase. Consistently, forced targeting of Kin4 to both SPBs delays mitotic exit even when the anaphase spindle is correctly aligned. Moreover, we present evidence that Spc72 has an additional function in SPOC regulation that is independent of the recruitment of Kin4. Thus, Spc72 provides a missing link between cytoplasmic MT function and components of the SPOC.
APA, Harvard, Vancouver, ISO, and other styles
49

Chauhan, Sujata, Subhasis Samanta, Nitin Sharma, Jitendra K. Thakur, Kamal Dev, and Anuradha Sourirajan. "Saccharomyces cerevisiae polo-like kinase, Cdc5 exhibits ATP-dependent Mg2+-enhanced kinase activity in vitro." Heliyon 5, no. 12 (December 2019): e03050. http://dx.doi.org/10.1016/j.heliyon.2019.e03050.

Full text
APA, Harvard, Vancouver, ISO, and other styles
50

Petersen, Janni, and Paul Nurse. "TOR signalling regulates mitotic commitment through the stress MAP kinase pathway and the Polo and Cdc2 kinases." Nature Cell Biology 9, no. 11 (October 21, 2007): 1263–72. http://dx.doi.org/10.1038/ncb1646.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography