Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
EMBO J ; 42(20): e114288, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37728253

ABSTRACT

Genome haploidization at meiosis depends on two consecutive nuclear divisions, which are controlled by an oscillatory system consisting of Cdk1-cyclin B and the APC/C bound to the Cdc20 activator. How the oscillator generates exactly two divisions has been unclear. We have studied this question in yeast where exit from meiosis involves accumulation of the APC/C activator Ama1 at meiosis II. We show that inactivation of the meiosis I-specific protein Spo13/MEIKIN results in a single-division meiosis due to premature activation of APC/CAma1 . In the wild type, Spo13 bound to the polo-like kinase Cdc5 prevents Ama1 synthesis at meiosis I by stabilizing the translational repressor Rim4. In addition, Cdc5-Spo13 inhibits the activity of Ama1 by converting the B-type cyclin Clb1 from a substrate to an inhibitor of Ama1. Cdc20-dependent degradation of Spo13 at anaphase I unleashes a feedback loop that increases Ama1's synthesis and activity, leading to irreversible exit from meiosis at the second division. Thus, by repressing the exit machinery at meiosis I, Cdc5-Spo13 ensures that cells undergo two divisions to produce haploid gametes.


Subject(s)
Saccharomyces cerevisiae Proteins , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Meiosis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Anaphase , Saccharomyces cerevisiae/metabolism , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , RNA-Binding Proteins/metabolism
2.
EMBO J ; 41(4): e109446, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35023198

ABSTRACT

Sexual reproduction requires genome haploidization by the two divisions of meiosis and a differentiation program to generate gametes. Here, we have investigated how sporulation, the yeast equivalent of gamete differentiation, is coordinated with progression through meiosis. Spore differentiation is initiated at metaphase II when a membrane-nucleating structure, called the meiotic plaque, is assembled at the centrosome. While all components of this structure accumulate already at entry into meiosis I, they cannot assemble because centrosomes are occupied by Spc72, the receptor of the γ-tubulin complex. Spc72 is removed from centrosomes by a pathway that depends on the polo-like kinase Cdc5 and the meiosis-specific kinase Ime2, which is unleashed by the degradation of Spo13/Meikin upon activation of the anaphase-promoting complex at anaphase I. Meiotic plaques are finally assembled upon reactivation of Cdk1 at entry into metaphase II. This unblocking-activation mechanism ensures that only single-copy genomes are packaged into spores and might serve as a paradigm for the regulation of other meiosis II-specific processes.


Subject(s)
Meiosis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Spores, Fungal/physiology , Cdc20 Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin B/metabolism , DNA-Binding Proteins/metabolism , Kinetochores/metabolism , Meiosis/physiology , Metaphase/physiology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/genetics , Spores, Fungal/cytology , Transcription Factors/metabolism
3.
EMBO J ; 40(7): e106812, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33644894

ABSTRACT

Genome haploidization involves sequential loss of cohesin from chromosome arms and centromeres during two meiotic divisions. At centromeres, cohesin's Rec8 subunit is protected from separase cleavage at meiosis I and then deprotected to allow its cleavage at meiosis II. Protection of centromeric cohesin by shugoshin-PP2A seems evolutionarily conserved. However, deprotection has been proposed to rely on spindle forces separating the Rec8 protector from cohesin at metaphase II in mammalian oocytes and on APC/C-dependent destruction of the protector at anaphase II in yeast. Here, we have activated APC/C in the absence of sister kinetochore biorientation at meiosis II in yeast and mouse oocytes, and find that bipolar spindle forces are dispensable for sister centromere separation in both systems. Furthermore, we show that at least in yeast, protection of Rec8 by shugoshin and inhibition of separase by securin are both required for the stability of centromeric cohesin at metaphase II. Our data imply that related mechanisms preserve the integrity of dyad chromosomes during the short metaphase II of yeast and the prolonged metaphase II arrest of mammalian oocytes.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Meiosis , Animals , Cells, Cultured , Female , Mice , Oocytes/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Cohesins
4.
Cell Cycle ; 16(12): 1145-1152, 2017 Jun 18.
Article in English | MEDLINE | ID: mdl-28514186

ABSTRACT

Cells undergoing meiosis produce haploid gametes through one round of DNA replication followed by 2 rounds of chromosome segregation. This requires that cohesin complexes, which establish sister chromatid cohesion during S phase, are removed in a stepwise manner. At meiosis I, the separase protease triggers the segregation of homologous chromosomes by cleaving cohesin's Rec8 subunit on chromosome arms. Cohesin persists at centromeres because the PP2A phosphatase, recruited by the shugoshin protein, dephosphorylates Rec8 and thereby protects it from cleavage. While chromatids disjoin upon cleavage of centromeric Rec8 at meiosis II, it was unclear how and when centromeric Rec8 is liberated from its protector PP2A. One proposal is that bipolar spindle forces separate PP2A from Rec8 as cells enter metaphase II. We show here that sister centromere biorientation is not sufficient to "deprotect" Rec8 at meiosis II in yeast. Instead, our data suggest that the ubiquitin-ligase APC/CCdc20 removes PP2A from centromeres by targeting for degradation the shugoshin Sgo1 and the kinase Mps1. This implies that Rec8 remains protected until entry into anaphase II when it is phosphorylated concurrently with the activation of separase. Here, we provide further support for this model and speculate on its relevance to mammalian oocytes.


Subject(s)
Cdc20 Proteins/physiology , Centromere/metabolism , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/enzymology , Animals , Cell Cycle Proteins/physiology , Centromere/genetics , Chromosomal Proteins, Non-Histone/physiology , Chromosome Segregation , Humans , Meiosis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Separase/physiology
5.
Dev Cell ; 40(1): 37-52, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28017619

ABSTRACT

Meiosis consists of DNA replication followed by two consecutive nuclear divisions and gametogenesis or spore formation. While meiosis I has been studied extensively, less is known about the regulation of meiosis II. Here we show that Hrr25, the conserved casein kinase 1δ of budding yeast, links three mutually independent key processes of meiosis II. First, Hrr25 induces nuclear division by priming centromeric cohesin for cleavage by separase. Hrr25 simultaneously phosphorylates Rec8, the cleavable subunit of cohesin, and removes from centromeres the cohesin protector composed of shugoshin and the phosphatase PP2A. Second, Hrr25 initiates the sporulation program by inducing the synthesis of membranes that engulf the emerging nuclei at anaphase II. Third, Hrr25 mediates exit from meiosis II by activating pathways that trigger the destruction of M-phase-promoting kinases. Thus, Hrr25 synchronizes formation of the single-copy genome with gamete differentiation and termination of meiosis.


Subject(s)
Casein Kinase I/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gametogenesis , Meiosis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Anaphase , Cell Nucleus/metabolism , Centromere/metabolism , Phosphorylation , Protein Phosphatase 2/metabolism , Proteolysis , Separase/metabolism , Spindle Apparatus/metabolism , Cohesins
6.
Curr Biol ; 26(24): R1272-R1274, 2016 12 19.
Article in English | MEDLINE | ID: mdl-27997836

ABSTRACT

Alternation between DNA replication in the mother cell (S phase) and equal partitioning of the replicated chromosomes to the daughter cells (M phase) during eukaryotic cell division is governed by switches that flip protein kinases on and off. New work reveals that the M-phase promoting kinase is opposed by a phosphatase that also participates in a bistable switching mechanism.


Subject(s)
Cell Division/physiology , Chromosomes/physiology , DNA Replication , Gene Expression Regulation, Enzymologic/physiology , Protein Kinases/metabolism , S Phase/physiology , Animals , Eukaryotic Cells , Protein Kinases/genetics
7.
Cell ; 151(3): 603-18, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23101628

ABSTRACT

Whereas proliferating cells enter M phase shortly after DNA replication, the first M phase of meiosis is preceded by an extended prophase in which homologous chromosomes undergo recombination. Exit from prophase I is controlled by the recombination checkpoint (RC), which, in yeast, represses the meiosis-specific transcription factor Ndt80 required for the expression of B-type cyclins and other M phase regulators. We show that an extended prophase I additionally requires the suppression of latent, mitotic cell-cycle controls by the anaphase-promoting complex (APC/C) and its meiosis-specific activator Ama1, which trigger the degradation of M phase regulators and Ndd1, a subunit of a mitotic transcription factor. ama1Δ mutants exit from prophase I prematurely and independently of the RC, which results in recombination defects and chromosome missegregation. Thus, control of prophase I by meiotic mechanisms depends on the suppression of the alternative, mitotic mechanisms by a meiosis-specific form of the APC/C.


Subject(s)
Cell Cycle Proteins/metabolism , Meiosis , Prophase , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Cdc20 Proteins , Chromosome Segregation , Chromosomes, Fungal/metabolism , DNA-Binding Proteins/metabolism , Metaphase , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Spindle Apparatus , Transcription Factors/metabolism
8.
Curr Biol ; 21(15): 1259-67, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21802300

ABSTRACT

BACKGROUND: The ways in which cells set the size of intracellular structures is an important but largely unsolved problem [1]. Early embryonic divisions pose special problems in this regard. Many checkpoints common in somatic cells are missing from these divisions, which are characterized by rapid reductions in cell size and short cell cycles [2]. Embryonic cells must therefore possess simple and robust mechanisms that allow the size of many of their intracellular structures to rapidly scale with cell size. RESULTS: Here, we study the mechanism by which one structure, the centrosome, scales in size during the early embryonic divisions of C. elegans. We show that centrosome size is directly related to cell size and is independent of lineage. Two findings suggest that the total amount of maternally supplied centrosome proteins could limit centrosome size. First, the combined volume of all centrosomes formed at any one time in the developing embryo is constant. Second, the total volume of centrosomes in any one cell is independent of centrosome number. By increasing the amount of centrosome proteins in the cell, we provide evidence that one component that limits centrosome size is the conserved pericentriolar material protein SPD-2 [3], which we show binds to and targets polo-like kinase 1 [3, 4] to centrosomes. CONCLUSIONS: We propose a limiting component hypothesis, in which the volume of the cell sets centrosome size by limiting the total amount of centrosome components. This idea could be a general mechanism for setting the size of intracellular organelles during development.


Subject(s)
Caenorhabditis elegans/embryology , Centrosome , Embryo, Nonmammalian/metabolism , Animals , Cell Cycle , Cell Size
9.
Dev Cell ; 18(3): 397-409, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20230747

ABSTRACT

During meiosis, two rounds of chromosome segregation after a single round of DNA replication produce haploid gametes from diploid precursors. At meiosis I, maternal and paternal kinetochores are pulled toward opposite poles, and chiasmata holding bivalent chromosomes together are resolved by cleavage of cohesin's alpha-kleisin subunit (Rec8) along chromosome arms. This creates dyad chromosomes containing a pair of chromatids joined solely by cohesin at centromeres that had resisted cleavage. The discovery that centromeric Rec8 is protected from separase during meiosis I by shugoshin/MEI-S332 proteins that bind PP2A phosphatase suggests that phosphorylation either of separase or cohesin may be necessary for Rec8 cleavage. We show here that multiple phosphorylation sites within Rec8 as well as two different kinases, casein kinase 1delta/epsilon (CK1delta/epsilon) and Dbf4-dependent Cdc7 kinase (DDK), are required for Rec8 cleavage and meiosis I nuclear division. Rec8 with phosphomimetic mutations is no longer protected from separase at centromeres and is cleaved even when the two kinases are inhibited. Our data suggest that PP2A protects centromeric cohesion by opposing CK1delta/epsilon- and DDK-dependent phosphorylation of Rec8.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Casein Kinase Idelta/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Endopeptidases/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Binding Sites , Casein Kinase 1 epsilon/genetics , Casein Kinase I/genetics , Casein Kinase I/metabolism , Casein Kinase Idelta/genetics , Cell Cycle Proteins/genetics , Centromere/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Endopeptidases/genetics , Genes, Fungal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Meiosis/genetics , Meiosis/physiology , Models, Biological , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Separase , Cohesins
10.
Cell ; 135(4): 662-78, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-19013276

ABSTRACT

Meiosis differs from mitosis in that DNA replication is followed by the segregation of homologous chromosomes but not sister chromatids. This depends on the formation of interhomolog connections through crossover recombination and on the attachment of sister kinetochores to microtubules emanating from the same spindle pole. We show that in yeast, the Dbf4-dependent Cdc7 kinase (DDK) provides a link between premeiotic S phase, recombination, and monopolar attachment. Independently from its established role in initiating DNA replication, DDK promotes double-strand break formation, the first step of recombination, and the recruitment of the monopolin complex to kinetochores, which is essential for monopolar attachment. DDK regulates monopolin localization together with the polo-kinase Cdc5 bound to Spo13, probably through phosphorylation of the monopolin subunit Lrs4. Thus, activation of DDK both initiates DNA replication and commits meiotic cells to reductional chromosome segregation in the first division of meiosis.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Meiosis , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Cell Cycle , Chromosomes/ultrastructure , DNA Replication , Gene Deletion , Kinetochores/metabolism , Kinetochores/ultrastructure , Microtubules/metabolism , Models, Biological , Models, Genetic , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Saccharomyces cerevisiae
11.
BMC Dev Biol ; 7: 19, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17374146

ABSTRACT

BACKGROUND: RNA interference coupled with videorecording of C. elegans embryos is a powerful method for identifying genes involved in cell division processes. Here we present a functional analysis of the gene B0511.9, previously identified as a candidate cell polarity gene in an RNAi videorecording screen of chromosome I embryonic lethal genes. RESULTS: Whereas weak RNAi inhibition of B0511.9 causes embryonic cell polarity defects, strong inhibition causes embryos to arrest in metaphase of meiosis I. The range of defects induced by RNAi of B0511.9 is strikingly similar to those displayed by mutants of anaphase-promoting complex/cyclosome (APC/C) components. Although similarity searches did not reveal any obvious homologue of B0511.9 in the non-redundant protein database, we found that the N-terminus shares a conserved sequence pattern with the N-terminus of the small budding yeast APC/C subunit Cdc26 and its orthologues from a variety of other organisms. Furthermore, we show that B0511.9 robustly complements the temperature-sensitive growth defect of a yeast cdc26Delta mutant. CONCLUSION: These data demonstrate that B0511.9 encodes the C. elegans APC/C subunit CDC-26.


Subject(s)
Caenorhabditis elegans/genetics , Cell Polarity/genetics , Genes, cdc , Meiosis/genetics , Saccharomyces cerevisiae/genetics , Anaphase-Promoting Complex-Cyclosome , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Embryo, Nonmammalian/cytology , Gene Expression Profiling , Genetic Complementation Test , Genetic Vectors , Phenotype , RNA Interference , RNA, Helminth/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Video Recording
12.
Cell ; 126(6): 1049-64, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-16990132

ABSTRACT

In meiosis, a single round of DNA replication is followed by two consecutive rounds of chromosome segregation, called meiosis I and II. Disjunction of maternal from paternal centromeres during meiosis I depends on the attachment of sister kinetochores to microtubules emanating from the same pole. In budding yeast, monopolar attachment requires recruitment to kinetochores of the monopolin complex. How monopolin promotes monopolar attachment was unclear, as its subunits are poorly conserved and lack similarities to proteins with known functions. We show here that the monopolin subunit Mam1 binds tightly to Hrr25, a highly conserved casein kinase 1 delta/epsilon (CK1delta/epsilon), and recruits it to meiosis I centromeres. Hrr25 kinase activity and Mam1 binding are both essential for monopolar attachment. Since CK1delta/epsilon activity is important for accurate chromosome segregation during meiosis I also in fission yeast, phosphorylation of kinetochore proteins by CK1delta/epsilon might be an evolutionary conserved process required for monopolar attachment.


Subject(s)
Casein Kinase I/metabolism , Chromosome Segregation/physiology , Kinetochores/metabolism , Meiosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Spindle Apparatus/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Casein Kinase I/genetics , Centromere/genetics , Centromere/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication/physiology , Kinetochores/ultrastructure , Protein Subunits/genetics , Protein Subunits/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/ultrastructure
13.
Cell ; 120(6): 773-88, 2005 Mar 25.
Article in English | MEDLINE | ID: mdl-15797379

ABSTRACT

Cohesion established between sister chromatids during pre-meiotic DNA replication mediates two rounds of chromosome segregation. The first division is preceded by an extended prophase wherein homologous chromosomes undergo recombination. The persistence of cohesion during prophase is essential for recombination and both meiotic divisions. Here we show that Mnd2, a subunit of the anaphase-promoting complex (APC/C) from budding yeast, is essential to prevent premature destruction of cohesion in meiosis. During S- and prophase, Mnd2 prevents activation of the APC/C by a meiosis-specific activator called Ama1. In cells lacking Mnd2 the APC/C-Ama1 enzyme triggers degradation of Pds1, which causes premature sister chromatid separation due to unrestrained separase activity. In vitro, Mnd2 inhibits ubiquitination of Pds1 by APC/C-Ama1 but not by other APC/C holo-enzymes. We conclude that chromosome segregation in meiosis depends on the selective inhibition of a meiosis-specific form of the APC/C.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosome Segregation/physiology , Meiosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase/genetics , Anaphase/physiology , Anaphase-Promoting Complex-Cyclosome , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , CDC28 Protein Kinase, S cerevisiae/genetics , CDC28 Protein Kinase, S cerevisiae/metabolism , Cdc20 Proteins , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromatids/metabolism , Chromosome Segregation/genetics , Endopeptidases/metabolism , Meiosis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Denaturation/physiology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Securin , Separase , Ubiquitin-Protein Ligase Complexes/genetics
14.
Dev Cell ; 8(3): 301-3, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15737925

ABSTRACT

Vertebrate eggs prevent parthenogenetic development by producing cytostatic factor (CSF), which blocks exit from metaphase of meiosis II until fertilization. CSF was never purified but recently suspected to inhibit the anaphase-promoting complex (APC), an ubiquitin ligase required for entry into anaphase. In a recent paper in Genes & Development, Schmidt et al. describe the Xenopus APC inhibitor Erp1, which seems to be the best candidate yet for the downstream effector of CSF activity.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Meiosis/physiology , Oocytes/cytology , Ubiquitin-Protein Ligase Complexes/metabolism , Xenopus Proteins/metabolism , Anaphase/physiology , Anaphase-Promoting Complex-Cyclosome , Animals , Calcium/metabolism , Female , Fertilization/physiology , Oocytes/metabolism , Proto-Oncogene Proteins c-mos/metabolism , Xenopus laevis/metabolism
15.
Curr Biol ; 14(24): 2183-96, 2004 Dec 29.
Article in English | MEDLINE | ID: mdl-15620645

ABSTRACT

BACKGROUND: Cells undergoing meiosis perform two consecutive divisions after a single round of DNA replication. During the first meiotic division, homologous chromosomes segregate to opposite poles. This is achieved by (1) the pairing of maternal and paternal chromosomes via recombination producing chiasmata, (2) coorientation of homologous chromosomes such that sister chromatids attach to the same spindle pole, and (3) resolution of chiasmata by proteolytic cleavage by separase of the meiotic-specific cohesin Rec8 along chromosome arms. Crucially, cohesin at centromeres is retained to allow sister centromeres to biorient at the second division. Little is known about how these meiosis I-specific events are regulated. RESULTS: Here, we show that Spo13, a centromere-associated protein produced exclusively during meiosis I, is required to prevent sister kinetochore biorientation by facilitating the recruitment of the monopolin complex to kinetochores. Spo13 is also required for the reaccumulation of securin, the persistence of centromeric cohesin during meiosis II, and the maintenance of a metaphase I arrest induced by downregulation of the APC/C activator CDC20. CONCLUSION: Spo13 is a key regulator of several meiosis I events. The presence of Spo13 at centromere-surrounding regions is consistent with the notion that it plays a direct role in both monopolin recruitment to centromeres during meiosis I and maintenance of centromeric cohesion between the meiotic divisions. Spo13 may also limit separase activity after the first division by ensuring securin reaccumulation and, in doing so, preventing precocious removal from chromatin of centromeric cohesin.


Subject(s)
Centromere/metabolism , Chromosome Segregation/physiology , Kinetochores/metabolism , Meiosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , ATP-Binding Cassette Transporters/metabolism , Blotting, Western , Cell Cycle Proteins/metabolism , Immunoprecipitation , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Securin , Yeasts
16.
Mol Cell Biol ; 24(8): 3562-76, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15060174

ABSTRACT

The anaphase-promoting complex (APC/C) is a large ubiquitin-protein ligase which controls progression through anaphase by triggering the degradation of cell cycle regulators such as securin and B-type cyclins. The APC/C is an unusually complex ligase containing at least 10 different, evolutionarily conserved components. In contrast to APC/C's role in cell cycle regulation little is known about the functions of individual subunits and how they might interact with each other. Here, we have analyzed Swm1/Apc13, a small subunit recently identified in the budding yeast complex. Database searches revealed proteins related to Swm1/Apc13 in various organisms including humans. Both the human and the fission yeast homologues are associated with APC/C subunits, and they complement the phenotype of an SWM1 deletion mutant of budding yeast. Swm1/Apc13 promotes the stable association with the APC/C of the essential subunits Cdc16 and Cdc27. Accordingly, Swm1/Apc13 is required for ubiquitin ligase activity in vitro and for the timely execution of APC/C-dependent cell cycle events in vivo.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Subunits/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Animals , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome , Cell Cycle/physiology , Chromatids/metabolism , DNA Polymerase III , Evolution, Molecular , Genetic Complementation Test , Humans , Meiosis/physiology , Molecular Sequence Data , Protein Subunits/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins , Sequence Alignment , Ubiquitin-Protein Ligase Complexes/chemistry , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligases
17.
Mol Cell ; 13(1): 2-3, 2004 Jan 16.
Article in English | MEDLINE | ID: mdl-14731388

ABSTRACT

Destruction boxes mark cyclin B and other proteins degraded in mitosis for ubiquitination by the anaphase-promoting complex (APC/C). In a paper in this issue of Molecular Cell, Yamano et al. show that destruction boxes directly bind to the APC/C in a cell cycle-regulated manner. Interestingly, this interaction does not require APC/C activators of the Cdc20 family, which were thought to be essential for recruiting substrates to the APC/C.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , Cyclin B/chemistry , Cyclin B/metabolism , Ubiquitin/metabolism , Animals , Cyclin B/genetics , Ligases , Mitosis , Mutation , Xenopus
18.
Mol Cell ; 12(1): 87-100, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12887895

ABSTRACT

The WD repeat protein Cdc20 is essential for progression through mitosis because it is required to activate ubiquitin ligation by the anaphase-promoting complex (APC/C). Here we show in yeast that Cdc20 binds to the CCT chaperonin, which is known as a folding machine for actin and tubulin. The CCT is required for Cdc20's ability to bind and activate the APC/C. In vivo, CCT is essential for Cdc20-dependent cell cycle events such as sister chromatid separation and exit from mitosis. The chaperonin is also required for the function of the Cdc20-related protein Cdh1, which activates the APC/C during G1. We propose that folding of the Cdc20 family of APC/C activators is an essential and evolutionary conserved function of the CCT chaperonin.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , Ligases/metabolism , Mitosis/genetics , Ubiquitin-Protein Ligase Complexes , Yeasts/metabolism , Adenosine Triphosphate/metabolism , Anaphase-Promoting Complex-Cyclosome , Cell Cycle Proteins/genetics , Chaperonin Containing TCP-1 , Chaperonins/genetics , Chromosome Segregation/genetics , Evolution, Molecular , Genes, cdc/physiology , Hydrolysis , Ligases/genetics , Protein Binding/physiology , Protein Folding , Protein Structure, Tertiary/genetics , Yeasts/genetics
SELECTION OF CITATIONS
SEARCH DETAIL