Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nature ; 580(7804): 542-547, 2020 04.
Article in English | MEDLINE | ID: mdl-32322059

ABSTRACT

Prolonged mitosis often results in apoptosis1. Shortened mitosis causes tumorigenic aneuploidy, but it is unclear whether it also activates the apoptotic machinery2. Separase, a cysteine protease and trigger of all eukaryotic anaphases, has a caspase-like catalytic domain but has not previously been associated with cell death3,4. Here we show that human cells that enter mitosis with already active separase rapidly undergo death in mitosis owing to direct cleavage of anti-apoptotic MCL1 and BCL-XL by separase. Cleavage not only prevents MCL1 and BCL-XL from sequestering pro-apoptotic BAK, but also converts them into active promoters of death in mitosis. Our data strongly suggest that the deadliest cleavage fragment, the C-terminal half of MCL1, forms BAK/BAX-like pores in the mitochondrial outer membrane. MCL1 and BCL-XL are turned into separase substrates only upon phosphorylation by NEK2A. Early mitotic degradation of this kinase is therefore crucial for preventing apoptosis upon scheduled activation of separase in metaphase. Speeding up mitosis by abrogation of the spindle assembly checkpoint results in a temporal overlap of the enzymatic activities of NEK2A and separase and consequently in cell death. We propose that NEK2A and separase jointly check on spindle assembly checkpoint integrity and eliminate cells that are prone to chromosome missegregation owing to accelerated progression through early mitosis.


Subject(s)
Apoptosis , Mitosis , Separase/metabolism , Animals , Cell Line , Cell Survival , Chromosome Segregation , Humans , M Phase Cell Cycle Checkpoints , Mice , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , NIMA-Related Kinases/metabolism , Phosphorylation , Substrate Specificity , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/metabolism
2.
Nature ; 580(7804): 536-541, 2020 04.
Article in English | MEDLINE | ID: mdl-32322060

ABSTRACT

Separation of eukaryotic sister chromatids during the cell cycle is timed by the spindle assembly checkpoint (SAC) and ultimately triggered when separase cleaves cohesion-mediating cohesin1-3. Silencing of the SAC during metaphase activates the ubiquitin ligase APC/C (anaphase-promoting complex, also known as the cyclosome) and results in the proteasomal destruction of the separase inhibitor securin1. In the absence of securin, mammalian chromosomes still segregate on schedule, but it is unclear how separase is regulated under these conditions4,5. Here we show that human shugoshin 2 (SGO2), an essential protector of meiotic cohesin with unknown functions in the soma6,7, is turned into a separase inhibitor upon association with SAC-activated MAD2. SGO2-MAD2 can functionally replace securin and sequesters most separase in securin-knockout cells. Acute loss of securin and SGO2, but not of either protein individually, resulted in separase deregulation associated with premature cohesin cleavage and cytotoxicity. Similar to securin8,9, SGO2 is a competitive inhibitor that uses a pseudo-substrate sequence to block the active site of separase. APC/C-dependent ubiquitylation and action of the AAA-ATPase TRIP13 in conjunction with the MAD2-specific adaptor p31comet liberate separase from SGO2-MAD2 in vitro. The latter mechanism facilitates a considerable degree of sister chromatid separation in securin-knockout cells that lack APC/C activity. Thus, our results identify an unexpected function of SGO2 in mitotically dividing cells and a mechanism of separase regulation that is independent of securin but still supervised by the SAC.


Subject(s)
Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/metabolism , Mad2 Proteins/metabolism , Securin , Separase/antagonists & inhibitors , ATPases Associated with Diverse Cellular Activities/metabolism , Anaphase-Promoting Complex-Cyclosome/metabolism , Cdc20 Proteins/metabolism , Cell Line , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Humans , Protein Binding , Securin/metabolism , Separase/metabolism , Cohesins
3.
Mol Cell ; 58(3): 495-506, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25921067

ABSTRACT

Ring-shaped cohesin keeps sister chromatids paired until cleavage of its Scc1/Rad21 subunit by separase triggers chromosome segregation in anaphase. Vertebrate separase is held inactive by mutually exclusive binding to securin or Cdk1-cyclin B1 and becomes unleashed only upon ubiquitin-dependent degradation of these regulators. Although most separase is usually found in association with securin, this anaphase inhibitor is dispensable for murine life while Cdk1-cyclin B1-dependent control of separase is essential. Here, we show that securin-independent inhibition of separase by Cdk1-cyclin B1 in early mitosis requires the phosphorylation-specific peptidyl-prolyl cis/trans isomerase Pin1. Furthermore, isomerization of previously securin-bound separase at the metaphase-to-anaphase transition renders it resistant to re-inhibition by residual securin. At the same time, isomerization also limits the half-life of separase's proteolytic activity, explaining how cohesin can be reloaded onto telophase chromatin in the absence of securin and cyclin B1 without being cleaved.


Subject(s)
Chromosome Segregation/genetics , Gene Expression Regulation, Enzymologic , Peptidylprolyl Isomerase/genetics , Separase/genetics , Anaphase/genetics , CDC2 Protein Kinase , Chromatids/genetics , Cyclin B1/chemistry , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , HEK293 Cells , Humans , Immunoblotting , Metaphase/genetics , Microscopy, Fluorescence , Mitosis/genetics , Models, Genetic , Models, Molecular , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/metabolism , Protein Binding , Protein Conformation , RNA Interference , Securin/genetics , Securin/metabolism , Separase/chemistry , Separase/metabolism
4.
EMBO J ; 37(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30305303

ABSTRACT

Separase halves eukaryotic chromosomes in M-phase by cleaving cohesin complexes holding sister chromatids together. Whether this essential protease functions also in interphase and/or impacts carcinogenesis remains largely unknown. Here, we show that mammalian separase is recruited to DNA double-strand breaks (DSBs) where it is activated to locally cleave cohesin and facilitate homology-directed repair (HDR). Inactivating phosphorylation of its NES, arginine methylation of its RG-repeats, and sumoylation redirect separase from the cytosol to DSBs. In vitro assays suggest that DNA damage response-relevant ATM, PRMT1, and Mms21 represent the corresponding kinase, methyltransferase, and SUMO ligase, respectively. SEPARASE heterozygosity not only debilitates HDR but also predisposes primary embryonic fibroblasts to neoplasia and mice to chemically induced skin cancer. Thus, tethering of separase to DSBs and confined cohesin cleavage promote DSB repair in G2 cells. Importantly, this conserved interphase function of separase protects mammalian cells from oncogenic transformation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA Breaks, Double-Stranded , Interphase , Neoplasm Proteins/metabolism , Recombinational DNA Repair , Separase/metabolism , Skin Neoplasms/enzymology , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enzyme Activation , HEK293 Cells , Humans , Ligases/genetics , Ligases/metabolism , Mice , Neoplasm Proteins/genetics , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Separase/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control
5.
J Cell Sci ; 131(11)2018 06 11.
Article in English | MEDLINE | ID: mdl-29724914

ABSTRACT

The DNA-embracing, ring-shaped multiprotein complex cohesin mediates sister chromatid cohesion and is stepwise displaced in mitosis by Wapl and separase (also known as ESPL1) to facilitate anaphase. Proper regulation of chromosome cohesion throughout meiosis is critical for preventing formation of aneuploid gametes, which are associated with trisomies and infertility in humans. Studying cohesion in meiocytes is complicated by their difficult experimental amenability and the absence of cohesin turnover. Here, we use cultured somatic cells to unravel fundamental aspects of meiotic cohesin. When expressed in Hek293 cells, the kleisin Rec8 displays no affinity for the peripheral cohesin subunits Stag1 or Stag2 and remains cytoplasmic. However, co-expression of Stag3 is sufficient for Rec8 to enter the nucleus, load onto chromatin, and functionally replace its mitotic counterpart Scc1 (also known as RAD21) during sister chromatid cohesion and dissolution. Rec8-Stag3 cohesin physically interacts with Pds5, Wapl and sororin (also known as CDCA5). Importantly, Rec8-Stag3 cohesin is shown to be susceptible to Wapl-dependent ring opening and sororin-mediated protection. These findings exemplify that our model system is suitable to rapidly generate testable predictions for important unresolved issues of meiotic cohesion regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Meiosis , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromatids/metabolism , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , DNA-Binding Proteins , HEK293 Cells , Humans , Nuclear Proteins/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Cohesins
6.
Mol Cell ; 44(1): 85-96, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21981920

ABSTRACT

Faithful transmission of genomic information requires tight spatiotemporal regulation of DNA replication factors. In the licensing step of DNA replication, CDT-1 is loaded onto chromatin to subsequently promote the recruitment of additional replication factors, including CDC-45 and GINS. During the elongation step, the CDC-45/GINS complex moves with the replication fork; however, it is largely unknown how its chromatin association is regulated. Here, we show that the chaperone-like ATPase CDC-48/p97 coordinates degradation of CDT-1 with release of the CDC-45/GINS complex. C. elegans embryos lacking CDC-48 or its cofactors UFD-1/NPL-4 accumulate CDT-1 on mitotic chromatin, indicating a critical role of CDC-48 in CDT-1 turnover. Strikingly, CDC-48(UFD-1/NPL-4)-deficient embryos show persistent chromatin association of CDC-45/GINS, which is a consequence of CDT-1 stabilization. Moreover, our data confirmed a similar regulation in Xenopus egg extracts, emphasizing a conserved coordination of licensing and elongation events during eukaryotic DNA replication by CDC-48/p97.


Subject(s)
Adenosine Triphosphatases/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , Ligases/metabolism , Xenopus Proteins/metabolism , Animals , Caenorhabditis elegans , Male , Mitosis , RNA Interference , Spermatozoa/metabolism , Two-Hybrid System Techniques , Ubiquitin/chemistry , Ubiquitin/metabolism , Valosin Containing Protein , Xenopus laevis
7.
EMBO J ; 33(10): 1134-47, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24781523

ABSTRACT

The universal triggering event of eukaryotic chromosome segregation is cleavage of centromeric cohesin by separase. Prior to anaphase, most separase is kept inactive by association with securin. Protein phosphatase 2A (PP2A) constitutes another binding partner of human separase, but the functional relevance of this interaction has remained enigmatic. We demonstrate that PP2A stabilizes separase-associated securin by dephosphorylation, while phosphorylation of free securin enhances its polyubiquitylation by the ubiquitin ligase APC/C and proteasomal degradation. Changing PP2A substrate phosphorylation sites to alanines slows degradation of free securin, delays separase activation, lengthens early anaphase, and results in anaphase bridges and DNA damage. In contrast, separase-associated securin is destabilized by introduction of phosphorylation-mimetic aspartates or extinction of separase-associated PP2A activity. G2- or prometaphase-arrested cells suffer from unscheduled activation of separase when endogenous securin is replaced by aspartate-mutant securin. Thus, PP2A-dependent stabilization of separase-associated securin prevents precocious activation of separase during checkpoint-mediated arrests with basal APC/C activity and increases the abruptness and fidelity of sister chromatid separation in anaphase.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Protein Phosphatase 2/metabolism , Securin/metabolism , Separase/metabolism , Anaphase-Promoting Complex-Cyclosome/genetics , HeLa Cells , Humans , Immunoprecipitation , Protein Phosphatase 2/genetics , Securin/genetics , Separase/genetics , Ubiquitination
8.
Biochim Biophys Acta ; 1858(9): 2035-2040, 2016 09.
Article in English | MEDLINE | ID: mdl-27287725

ABSTRACT

Organelles in eukaryotic cells often have complex shapes that deviate significantly from simple spheres. A prime example is the endoplasmic reticulum (ER) that forms an extensive network of membrane tubules in many mammalian cell types and in reconstitution assays in vitro. Despite the successful hunt for molecular determinants of ER shape we are still far from having a comprehensive understanding of ER network morphogenesis. Here, we have studied the hitherto neglected influence of the host substrate when reconstituting ER networks in vitro as compared to ER networks in vivo. In culture cells we observed cytoplasm-spanning ER networks with tubules being connected almost exclusively by three-way junctions and segment lengths being narrowly distributed around a mean length of about 1µm. In contrast, networks reconstituted from purified ER microsomes on flat glass or gel substrates of varying stiffness showed significantly broader length distributions with an up to fourfold larger mean length. Self-assembly of ER microsomes on small oil droplets, however, yielded networks that resembled more closely the native ER network of mammalian cells. We conclude from these observations that the ER microsomes' inherent self-assembly capacity is sufficient to support network formation with a native geometry if the influence of the host substrate's surface chemistry becomes negligible. We hypothesize that under these conditions the networks' preference for three-way junctions follows from creating 'starfish-shaped' vesicles when ER microsomes with a protein-induced spontaneous curvature undergo fusion.


Subject(s)
Cytoplasm/chemistry , Endoplasmic Reticulum/chemistry , HeLa Cells , Humans
9.
EMBO J ; 32(5): 666-76, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23361318

ABSTRACT

Faithful transmission of chromosomes during eukaryotic cell division requires sister chromatids to be paired from their generation in S phase until their separation in M phase. Cohesion is mediated by the cohesin complex, whose Smc1, Smc3 and Scc1 subunits form a tripartite ring that entraps both DNA double strands. Whereas centromeric cohesin is removed in late metaphase by Scc1 cleavage, metazoan cohesin at chromosome arms is displaced already in prophase by proteolysis-independent signalling. Which of the three gates is triggered by the prophase pathway to open has remained enigmatic. Here, we show that displacement of human cohesin from early mitotic chromosomes requires dissociation of Smc3 from Scc1 but no opening of the other two gates. In contrast, loading of human cohesin onto chromatin in telophase occurs through the Smc1-Smc3 hinge. We propose that the use of differently regulated gates for loading and release facilitates unidirectionality of DNA's entry into and exit from the cohesin ring.


Subject(s)
Cell Cycle Proteins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Prophase/physiology , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cells, Cultured , Chondroitin Sulfate Proteoglycans/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human/metabolism , DNA/genetics , DNA-Binding Proteins , Humans , Immunoprecipitation , Immunosuppressive Agents/pharmacology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Microscopy, Fluorescence , Nuclear Proteins/genetics , Phosphoproteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Cohesins
10.
J Biol Chem ; 290(12): 8002-10, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25659430

ABSTRACT

Sister chromatid cohesion is established during replication by entrapment of both dsDNAs within the cohesin ring complex. It is dissolved in anaphase when separase, a giant cysteine endopeptidase, cleaves the Scc1/Rad21 subunit of cohesin, thereby triggering chromosome segregation. Separase is held inactive by association with securin until this anaphase inhibitor is destroyed at the metaphase-to-anaphase transition by ubiquitin-dependent degradation. The relevant ubiquitin ligase, the anaphase-promoting complex/cyclosome, also targets cyclin B1, thereby causing inactivation of Cdk1 and mitotic exit. Although separase is essential, securin knock-out mice are surprisingly viable and fertile. Capitalizing on our previous finding that Cdk1-cyclin B1 can also bind and inhibit separase, we investigated whether this kinase might be suitable to maintain faithful timing and execution of anaphase in the absence of securin. We found that, similar to securin, Cdk1-cyclin B1 regulates separase in both a positive and negative manner. Although securin associates with nascent separase to co-translationally assist proper folding, Cdk1-cyclin B1 acts on native state separase. Upon entry into mitosis, Cdk1-cyclin B1-dependent phosphorylation of Ser-1126 renders separase prone to inactivation by aggregation/precipitation. Stable association of Cdk1-cyclin B1 with phosphorylated separase counteracts this tendency and stabilizes separase in an inhibited yet activatable state. These opposing effects are suited to prevent premature cleavage of cohesin in early mitosis while ensuring timely activation of separase by anaphase-promoting complex/cyclosome-dependent degradation of cyclin B1. Coupling sister chromatid separation with subsequent exit from mitosis by this simplified mode might have been the common scheme of mitotic control prior to the evolution of securin.


Subject(s)
Cyclin B1/physiology , Cyclin-Dependent Kinases/physiology , Securin/physiology , Separase/physiology , Base Sequence , CDC2 Protein Kinase , DNA Primers , Flow Cytometry , HEK293 Cells , Humans , RNA Interference
11.
Mol Cell ; 31(3): 438-48, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18691976

ABSTRACT

Protein kinases are pivotal regulators of cell signaling that modulate each other's functions and activities through site-specific phosphorylation events. These key regulatory modifications have not been studied comprehensively, because low cellular abundance of kinases has resulted in their underrepresentation in previous phosphoproteome studies. Here, we combine kinase-selective affinity purification with quantitative mass spectrometry to analyze the cell-cycle regulation of protein kinases. This proteomics approach enabled us to quantify 219 protein kinases from S and M phase-arrested human cancer cells. We identified more than 1000 phosphorylation sites on protein kinases. Intriguingly, half of all kinase phosphopeptides were upregulated in mitosis. Our data reveal numerous unknown M phase-induced phosphorylation sites on kinases with established mitotic functions. We also find potential phosphorylation networks involving many protein kinases not previously implicated in mitotic progression. These results provide a vastly extended knowledge base for functional studies on kinases and their regulation through site-specific phosphorylation.


Subject(s)
Cell Cycle , Phosphoproteins/analysis , Phosphotransferases/metabolism , Proteomics , Amino Acid Sequence , Enzyme Activation , HeLa Cells , Humans , Mitosis , Molecular Sequence Data , Phosphopeptides/analysis , Phosphorylation , Phosphotransferases/chemistry , S Phase , Substrate Specificity
12.
EMBO J ; 30(14): 2868-80, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21666598

ABSTRACT

Mammalian centromeric cohesin is protected from phosphorylation-dependent displacement in mitotic prophase by shugoshin-1 (Sgo1), while shugoshin-2 (Sgo2) protects cohesin from separase-dependent cleavage in meiosis I. In higher eukaryotes, progression and faithful execution of both mitosis and meiosis are controlled by the spindle assembly checkpoint, which delays anaphase onset until chromosomes have achieved proper attachment to microtubules. According to the so-called template model, Mad1-Mad2 complexes at unattached kinetochores instruct conformational change of soluble Mad2, thus catalysing Mad2 binding to its target Cdc20. Here, we show that human Sgo2, but not Sgo1, specifically interacts with Mad2 in a manner that strongly resembles the interactions of Mad2 with Mad1 or Cdc20. Sgo2 contains a Mad1/Cdc20-like Mad2-interaction motif and competes with Mad1 and Cdc20 for binding to Mad2. NMR and biochemical analyses show that shugoshin binding induces similar conformational changes in Mad2 as do Mad1 or Cdc20. Mad2 binding regulates fine-tuning of Sgo2's sub-centromeric localization. Mad2 binding is conserved in the only known Xenopus laevis shugoshin homologue and, compatible with a putative meiotic function, the interaction occurs in oocytes.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Meiosis/physiology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium-Binding Proteins/genetics , Cdc20 Proteins , Cell Cycle Proteins/genetics , Cells, Cultured , Centromere/physiology , Female , Humans , Kinetochores/physiology , Mad2 Proteins , Molecular Sequence Data , Nuclear Proteins/genetics , Oocytes/cytology , Oocytes/metabolism , Protein Conformation , Repressor Proteins/genetics , Sequence Homology, Amino Acid , Spindle Apparatus , Xenopus laevis/metabolism
13.
Nat Cell Biol ; 8(9): 1035-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16906143

ABSTRACT

Separase not only triggers anaphase of meiosis I by proteolytic cleavage of cohesin on chromosome arms, but in vitro vertebrate separase also acts as a direct inhibitor of cyclin-dependent kinase 1 (Cdk1) on liberation from the inhibitory protein, securin. Blocking separase-Cdk1 complex formation by microinjection of anti-separase antibodies prevents polar-body extrusion in vertebrate oocytes. Importantly, proper meiotic maturation is rescued by chemical inhibition of Cdk1 or expression of Cdk1-binding separase fragments lacking cohesin-cleaving activity.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Endopeptidases/metabolism , Meiosis/physiology , Oocytes/physiology , Animals , CDC2 Protein Kinase/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line , Endopeptidases/genetics , Female , Histones/metabolism , Humans , Mice , Neoplasm Proteins/metabolism , Oocytes/metabolism , Securin , Separase , Xenopus
14.
EMBO Rep ; 12(5): 436-43, 2011 May.
Article in English | MEDLINE | ID: mdl-21399619

ABSTRACT

Mature Xenopus oocytes are arrested in meiosis by the activity of XErp1/Emi2, an inhibitor of the ubiquitin-ligase anaphase-promoting complex/cyclosome (APC/C). On fertilization, XErp1 is degraded, resulting in APC/C activation and the consequent degradation of cell-cycle regulators and exit from meiosis. In this study, we show that a modest increase in the activity of the ubiquitin-conjugating enzyme UbcX overrides the meiotic arrest in an APC/C-dependent reaction. Intriguingly, XErp1 remains stable in these conditions. We found that UbcX causes the ubiquitylation of XErp1, followed by its dissociation from the APC/C. Our data support the idea that ubiquitylation regulates the APC/C-inhibitory activity of XErp1.


Subject(s)
F-Box Proteins/metabolism , Meiosis/physiology , Oocytes/metabolism , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Xenopus Proteins/metabolism , Xenopus/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Blotting, Western , Plasmids/genetics , Ubiquitination , Xenopus/physiology
15.
JCI Insight ; 8(22)2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37796616

ABSTRACT

MAD2L1BP-encoded p31comet mediates Trip13-dependent disassembly of Mad2- and Rev7-containing complexes and, through this antagonism, promotes timely spindle assembly checkpoint (SAC) silencing, faithful chromosome segregation, insulin signaling, and homology-directed repair (HDR) of DNA double-strand breaks. We identified a homozygous MAD2L1BP nonsense variant, R253*, in 2 siblings with microcephaly, epileptic encephalopathy, and juvenile granulosa cell tumors of ovary and testis. Patient-derived cells exhibited high-grade mosaic variegated aneuploidy, slowed-down proliferation, and instability of truncated p31comet mRNA and protein. Corresponding recombinant p31comet was defective in Trip13, Mad2, and Rev7 binding and unable to support SAC silencing or HDR. Furthermore, C-terminal truncation abrogated an identified interaction of p31comet with tp53. Another homozygous truncation, R227*, detected in an early-deceased patient with low-level aneuploidy, severe epileptic encephalopathy, and frequent blood glucose elevations, likely corresponds to complete loss of function, as in Mad2l1bp-/- mice. Thus, human mutations of p31comet are linked to aneuploidy and tumor predisposition.


Subject(s)
Brain Diseases , Granulosa Cell Tumor , Ovarian Neoplasms , Female , Humans , Animals , Mice , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Granulosa Cell Tumor/genetics , Mutation , Aneuploidy
16.
J Cell Sci ; 123(Pt 5): 806-13, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20144989

ABSTRACT

Sister chromatid cohesion is mediated by DNA catenation and proteinaceous cohesin complexes. The recent visualization of PICH (Plk1-interacting checkpoint helicase)-coated DNA threads in anaphase cells raises new questions as to the role of DNA catenation and its regulation in time and space. In the present study we show that persistent DNA catenation induced by inhibition of Topoisomerase-IIalpha can contribute to sister chromatid cohesion in the absence of cohesin complexes and that resolution of catenation is essential for abscission. Furthermore, we use an in vitro chromatid separation assay to investigate the temporal and functional relationship between cohesin removal and Topoisomerase-IIalpha-mediated decatenation. Our data suggest that centromere decatenation can occur only after separase activation and cohesin removal, providing a plausible explanation for the persistence of centromere threads after anaphase onset.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Division/physiology , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA, Catenated/metabolism , DNA/metabolism , Anaphase/genetics , Anaphase/physiology , Cell Division/genetics , Cell Line , Cell Line, Tumor , Centromere/genetics , Chromatids/genetics , Chromatids/metabolism , DNA/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA, Catenated/genetics , Humans , Cohesins
17.
Nat Struct Mol Biol ; 12(6): 552-3, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15880121

ABSTRACT

After the degradation of its inhibitor securin, separase initiates chromosome segregation during the metaphase-to-anaphase transition by cleaving cohesin. Here we present a density map at a resolution of 25 A of negatively stained separase-securin complex. Based on labeling data and sequence analysis, we propose a model for the structure of separase, consisting of 26 ARM repeats, an unstructured region of 280 residues and two caspase-like domains, with securin binding to the ARM repeats.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Cell Cycle Proteins/ultrastructure , Endopeptidases/ultrastructure , Humans , Microscopy, Electron , Models, Molecular , Neoplasm Proteins/ultrastructure , Protein Conformation , Protein Folding , Securin , Separase
18.
Dev Biol ; 321(2): 379-86, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18639540

ABSTRACT

Mammalian eggs remain arrested at metaphase of the second meiotic division (metII) for an indeterminate time before fertilization. During this period, which can last several hours, the continued attachment of sister chromatids is thought to be achieved by inhibition of the protease separase. Separase is known to be inhibited by binding either securin or Maturation (M-Phase)-Promoting Factor, a heterodimer of CDK1/cyclin B1. However, the relative contribution of securin and CDK/cyclin B1 to sister chromatid attachment during metII arrest has not been assessed. Although there are conditions in which either CDK1/cyclinB1 activity or securin can prevent sister chromatid disjunction, principally by overexpression of non-degradable cyclin B1 or securin, we find here that separase activity is primarily regulated by securin and not CDK1/cyclin B1. Thus the CDK1 inhibitor roscovitine and an antibody we designed to block the interaction of CDK1/cyclin B1 with separase, both failed to induce sister disjunction. In contrast, securin morpholino knockdown specifically induced loss of sister attachment, that could be restored by securin cRNA rescue. During metII arrest separase appears primarily regulated by securin binding, not CDK1/cyclin B1.


Subject(s)
Carrier Proteins/metabolism , Chromatids/metabolism , Meiosis/physiology , Ovum/cytology , Animals , Blotting, Western , Cell Cycle Proteins/metabolism , Cytogenetic Analysis , Endopeptidases/metabolism , Mice , Oligonucleotides , Securin , Separase
19.
ACS Chem Biol ; 14(10): 2155-2159, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31553567

ABSTRACT

Separase, a cysteine protease of the CD clan, triggers chromosome segregation during mitosis by cleaving the cohesin ring entrapping the two sister chromatids. Deregulated separase activity is associated with aneuploidy, a hallmark of most human cancers. In fact, separase is highly overexpressed in many solid cancers, making it an attractive chemotherapeutic target. To identify small molecules capable of inhibiting separase in its complex cellular environment, we established a highly sensitive assay to quantify separase activity in cells and screened a 51 009-member library for separase inhibitors. In vitro assays confirmed that the identified compounds efficiently inhibited separase, while not affecting caspase-1, another CD-clan protease structurally related to separase. Importantly, HeLa cells with compromised separase activity displayed severe chromosome segregation defects upon compound treatment, confirming that the identified inhibitors are bioactive in tumor tissue culture cells. Structure-activity relationship studies succeeded in the optimization of the most promising inhibitor. Overall, this study demonstrates the feasibility of identifying separase-specific inhibitors, which serve as promising lead compounds for the development of clinically relevant separase inhibiting drugs.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Separase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Chromosome Segregation/drug effects , Enzyme Assays , HeLa Cells , Humans
20.
PLoS Biol ; 3(12): e416, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16292982

ABSTRACT

Abnormalities of chromosome number are frequently observed in cancers. The mechanisms regulating chromosome segregation in human cells are therefore of great interest. Recently it has been reported that human cells without an hSecurin gene lose chromosomes at a high frequency. Here we show that, after hSecurin knockout through homologous recombination, chromosome losses are only a short, transient effect. After a few passages hSecurin(-/-) cells became chromosomally stable and executed mitoses normally. This was unexpected, as the securin loss resulted in a persisting reduction of the sister-separating protease separase and inefficient cleavage of the cohesin subunit Scc1. Our data demonstrate that securin is dispensable for chromosomal stability in human cells. We propose that human cells possess efficient mechanisms to compensate for the loss of genes involved in chromosome segregation.


Subject(s)
Chromosomal Instability/genetics , Neoplasm Proteins/metabolism , Anaphase , Cell Cycle Proteins/metabolism , Cell Line , Chromosomal Proteins, Non-Histone , Endopeptidases/metabolism , Humans , In Situ Hybridization, Fluorescence , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Securin , Separase
SELECTION OF CITATIONS
SEARCH DETAIL