Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Semin Cell Dev Biol ; 117: 52-61, 2021 09.
Article in English | MEDLINE | ID: mdl-34127384

ABSTRACT

The establishment of a metaphase plate in which all chromosomes are attached to mitotic spindle microtubules and aligned at the cell equator is required for faithful chromosome segregation in metazoans. The achievement of this configuration relies on the precise coordination between several concurrent mechanisms that start upon nuclear envelope breakdown, mediate chromosome capture at their kinetochores during mitotic spindle assembly and culminate with the congression of all chromosomes to the spindle equator. This period is called 'prometaphase'. Because the nature of chromosome capture by mitotic spindle microtubules is error prone, the cell is provided of error correction mechanisms that sense and correct most erroneous kinetochore-microtubule attachments before committing to separate sister chromatids in anaphase. In this review, aimed for newcomers in the field, more than providing an exhaustive mechanistic coverage of each and every concurrent mechanism taking place during prometaphase, we provide an integrative overview of these processes that ultimately promote the subsequent faithful segregation of chromosomes during mitosis.


Subject(s)
Mitosis/physiology , Prometaphase/physiology , Humans , Spindle Apparatus/metabolism
2.
Mol Biol Cell ; 32(10): 1020-1032, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33788584

ABSTRACT

In prophase of meiosis I, homologous chromosomes pair and become connected by cross-overs. Chiasmata, the connections formed by cross-overs, enable the chromosome pair, called a bivalent, to attach as a single unit to the spindle. When the meiotic spindle forms in prometaphase, most bivalents are associated with one spindle pole and then go through a series of oscillations on the spindle, attaching to and detaching from microtubules until the partners of the bivalent become bioriented-attached to microtubules from opposite sides of the spindle. The conserved kinase, Mps1, is essential for the bivalents to be pulled by microtubules across the spindle in prometaphase. Here we show that MPS1 is needed for efficient triggering of the migration of microtubule-attached kinetochores toward the poles and promotes microtubule depolymerization. Our data support the model Mps1 acts at the kinetochore to coordinate the successful attachment of a microtubule and the triggering of microtubule depolymerization to then move the chromosome.


Subject(s)
Chromosomes/physiology , Prometaphase/physiology , Protein Serine-Threonine Kinases/physiology , Saccharomyces cerevisiae Proteins/physiology , Cell Polarity , Chromosome Pairing , Kinetochores/physiology , Microtubules/physiology , Mutation , Prometaphase/genetics , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomycetales
3.
Chromosoma ; 127(4): 461-473, 2018 12.
Article in English | MEDLINE | ID: mdl-29968190

ABSTRACT

Formation of stable kinetochore-microtubule attachments is essential for accurate chromosome segregation in human cells and depends on the NDC80 complex. We recently showed that Chmp4c, an endosomal sorting complex required for transport protein involved in membrane remodelling, localises to prometaphase kinetochores and promotes cold-stable kinetochore microtubules, faithful chromosome alignment and segregation. In the present study, we show that Chmp4c associates with the NDC80 components Hec1 and Nuf2 and is required for optimal NDC80 stability and Hec1-Nuf2 localisation to kinetochores in prometaphase. However, Chmp4c-depletion does not cause a gross disassembly of outer or inner kinetochore complexes. Conversely, Nuf2 is required for Chmp4c kinetochore targeting. Constitutive Chmp4c kinetochore tethering partially rescues cold-stable microtubule polymers in cells depleted of the endogenous Nuf2, showing that Chmp4c also contributes to kinetochore-microtubule stability independently of regulating Hec1 and Nuf2 localisation. Chmp4c interacts with tubulin in cell extracts, and binds and bundles microtubules in vitro through its highly basic N-terminal region (amino acids 1-77). Furthermore, the N-terminal region of Chmp4c is required for cold-stable kinetochore microtubules and efficient chromosome alignment. We propose that Chmp4c promotes stable kinetochore-microtubule attachments by regulating Hec1-Nuf2 localisation to kinetochores in prometaphase and by binding to spindle microtubules. These results identify Chmp4c as a novel protein that regulates kinetochore-microtubule interactions to promote accurate chromosome segregation in human cells.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation , Cytoskeletal Proteins , Endosomal Sorting Complexes Required for Transport/genetics , HeLa Cells , Humans , Nuclear Proteins/metabolism , Prometaphase/physiology
4.
Sci Rep ; 8(1): 3888, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29497093

ABSTRACT

Faithful chromosome segregation is ensured by the establishment of bi-orientation; the attachment of sister kinetochores to the end of microtubules extending from opposite spindle poles. In addition, kinetochores can also attach to lateral surfaces of microtubules; called lateral attachment, which plays a role in chromosome capture and transport. However, molecular basis and biological significance of lateral attachment are not fully understood. We have addressed these questions by focusing on the prometaphase rosette, a typical chromosome configuration in early prometaphase. We found that kinetochores form uniform lateral attachments in the prometaphase rosette. Many transient kinetochore components are maximally enriched, in an Aurora B activity-dependent manner, when the prometaphase rosette is formed. We revealed that rosette formation is driven by rapid poleward motion of dynein, but can occur even in its absence, through slow kinetochore movements caused by microtubule depolymerization that is supposedly dependent on kinetochore tethering at microtubule ends by CENP-E. We also found that chromosome connection to microtubules is extensively lost when lateral attachment is perturbed in cells defective in end-on attachment. Our findings demonstrate that lateral attachment is an important intermediate in bi-orientation establishment and chromosome alignment, playing a crucial role in incorporating chromosomes into the nascent spindle.


Subject(s)
Chromosome Segregation/physiology , Kinetochores/physiology , Microtubules/physiology , Dyneins/metabolism , HeLa Cells , Humans , Prometaphase/physiology , Rosette Formation/methods , Spindle Apparatus/metabolism , Spindle Apparatus/physiology
5.
J Vis Exp ; (130)2017 12 06.
Article in English | MEDLINE | ID: mdl-29286418

ABSTRACT

In humans, chromosome segregation errors in oocytes are responsible for the majority of miscarriages and birth defects. Moreover, as women age, their risk of conceiving an aneuploid fetus increases dramatically and this phenomenon is known as the maternal age effect. One requirement for accurate chromosome segregation during the meiotic divisions is maintenance of sister chromatid cohesion during the extended prophase period that oocytes experience. Cytological evidence in both humans and model organisms suggests that meiotic cohesion deteriorates during the aging process. In addition, segregation errors in human oocytes are most prevalent during meiosis I, consistent with premature loss of arm cohesion. The use of model organisms is critical for unraveling the mechanisms that underlie age-dependent loss of cohesion. Drosophila melanogaster offers several advantages for studying the regulation of meiotic cohesion in oocytes. However, until recently, only genetic tests were available to assay for loss of arm cohesion in oocytes of different genotypes or under different experimental conditions. Here, a detailed protocol is provided for using fluorescence in situ hybridization (FISH) to directly visualize defects in arm cohesion in prometaphase I and metaphase I arrested Drosophila oocytes. By generating a FISH probe that hybridizes to the distal arm of the X chromosome and collecting confocal Z stacks, a researcher can visualize the number of individual FISH signals in three dimensions and determine whether sister chromatid arms are separated. The procedure outlined makes it possible to quantify arm cohesion defects in hundreds of Drosophila oocytes. As such, this method provides an important tool for investigating the mechanisms that contribute to cohesion maintenance as well as the factors that lead to its demise during the aging process.


Subject(s)
Drosophila/physiology , In Situ Hybridization, Fluorescence/methods , Oocytes/cytology , Animals , Drosophila/cytology , Female , Humans , Metaphase/physiology , Prometaphase/physiology
6.
Nat Cell Biol ; 18(6): 692-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27136266

ABSTRACT

The formation of mitotic chromosomes requires both compaction of chromatin and the resolution of replicated sister chromatids. Compaction occurs during mitotic prophase and prometaphase, and in prophase relies on the activity of condensin II complexes. Exactly when and how sister chromatid resolution occurs has been largely unknown, as has its molecular requirements. Here, we established a method to visualize sister resolution by sequential replication labelling with two distinct nucleotide derivatives. Quantitative three-dimensional imaging then allowed us to measure the resolution of sister chromatids throughout mitosis by calculating their non-overlapping volume within the whole chromosome. Unexpectedly, we found that sister chromatid resolution starts already at the beginning of prophase, proceeds concomitantly with chromatin compaction and is largely completed by the end of prophase. Sister chromatid resolution was abolished by inhibition of topoisomerase IIα and by depleting or preventing mitotic activation of condensin II, whereas blocking cohesin dissociation from chromosomes had little effect. Mitotic sister chromatid resolution is thus an intrinsic part of mitotic chromosome formation in prophase that relies largely on DNA decatenation and shares the molecular requirement for condensin II with prophase compaction.


Subject(s)
Chromatids/metabolism , Mitosis/physiology , Prometaphase/physiology , Prophase/physiology , Adenosine Triphosphatases/metabolism , Antigens, Neoplasm/metabolism , Cell Line , DNA Replication/physiology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Humans , Imaging, Three-Dimensional/methods , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism
7.
Cell Cycle ; 13(11): 1663-70, 2014.
Article in English | MEDLINE | ID: mdl-24786771

ABSTRACT

Heterochromatin protein 1α (HP1α), a bona fide factor of silent chromatin, is required for establishing as well as maintaining the higher-order chromatin structure in eukaryotes. HP1α is decorated with several post-translational modifications, and many of these are critical for its cellular functions. HP1α is heavily phosphorylated; however, its physiological relevance had remained to be completely understood. We have recently demonstrated that human HP1α is a mitotic target for NDR kinase, and the phosphorylation at the hinge region of HP1α at the G 2/M phase of the cell cycle is crucial for mitotic progression and Sgo1 loading at mitotic centromeres (Chakraborty et al., 2014). We now demonstrate that the dephosphorylation of HP1α within its hinge domain occurs during mitosis, specifically soon after prometaphase. In the absence of the hinge-specific HP1α phosphorylation, either as a consequence of depleting NDR1 or in cells expressing a non-phosphorylatable HP1α mutant, the cells arrest in prometaphase with several mitotic defects. In this study we show that NDR1-depleted cells expressing hinge-specific phosphomimetic HP1α mutant rescues the prometaphase arrest but displays defects in mitotic exit, suggesting that the dephosphorylation of HP1α is required for the completion of cytokinesis. Taken together, our results reveal that the phosphorylation-dephosphorylation cycle of HP1α orchestrates accurate progression of cells through mitosis.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Mitosis/physiology , Cell Cycle Checkpoints/physiology , Cell Line , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Flow Cytometry , Humans , Phosphorylation , Prometaphase/physiology , Protein Serine-Threonine Kinases/metabolism
8.
Cell Cycle ; 13(8): 1327-34, 2014.
Article in English | MEDLINE | ID: mdl-24621506

ABSTRACT

Faithful chromosome segregation during meiosis is indispensable to prevent birth defects and infertility. Canonical genetic manipulations have not been very useful for studying meiosis II, since mutations of genes involved in cell cycle regulation or chromosome segregation may affect meiosis I, making interpretations of any defects observed in meiosis II complicated. Here we present a powerful strategy to dissect meiosis I and meiosis II, using chemical inhibitors in genetically tractable model organism fission yeast (Schizosaccharomyces pombe). As various chemical probes are not active in fission yeast, mainly due to an effective multidrug resistance (MDR) response, we have recently developed a drug-hypersensitive MDR-sup strain by suppression of the key genes responsible for MDR response. We further developed the MDR-supML (marker-less) strain by deleting 7 MDR genes without commonly used antibiotic markers. The new strain makes fluorescent tagging and gene deletion much simpler, which enables effective protein visualization in varied genetic backgrounds. Using the MDR-supML strain with chemical inhibitors and live cell fluorescence microscopy, we established cell cycle arrest at meiosis I and meiosis II and examined Aurora-dependent spindle assembly checkpoint (SAC) regulation during meiosis. We found that Aurora B/Ark1 kinase activity is required for recruitment of Bub1, an essential SAC kinase, to unattached kinetochore in prometaphase I and prometaphase II as in mitosis. Thus, Aurora's role in SAC activation is likely conserved in mitosis, meiosis I, and meiosis II. Together, our MDR-supML strain will be useful to dissect complex molecular mechanisms in mitosis and 2 successive meiotic divisions.


Subject(s)
Meiosis/physiology , Schizosaccharomyces/physiology , Antifungal Agents/pharmacology , Aurora Kinases/metabolism , Boronic Acids/pharmacology , Bortezomib , Brefeldin A/pharmacology , Cycloheximide/pharmacology , Drug Resistance, Multiple, Fungal , M Phase Cell Cycle Checkpoints/physiology , Prometaphase/physiology , Protein Serine-Threonine Kinases/metabolism , Pyrazines/pharmacology , Schizosaccharomyces/drug effects , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/metabolism
9.
EMBO J ; 32(2): 303-14, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23288039

ABSTRACT

The Anaphase Promoting Complex/Cyclosome (APC/C) in complex with its co-activator Cdc20 is responsible for targeting proteins for ubiquitin-mediated degradation during mitosis. The activity of APC/C-Cdc20 is inhibited during prometaphase by the Spindle Assembly Checkpoint (SAC) yet certain substrates escape this inhibition. Nek2A degradation during prometaphase depends on direct binding of Nek2A to the APC/C via a C-terminal MR dipeptide but whether this motif alone is sufficient is not clear. Here, we identify Kif18A as a novel APC/C-Cdc20 substrate and show that Kif18A degradation depends on a C-terminal LR motif. However in contrast to Nek2A, Kif18A is not degraded until anaphase showing that additional mechanisms contribute to Nek2A degradation. We find that dimerization via the leucine zipper, in combination with the MR motif, is required for stable Nek2A binding to and ubiquitination by the APC/C. Nek2A and the mitotic checkpoint complex (MCC) have an overlap in APC/C subunit requirements for binding and we propose that Nek2A binds with high affinity to apo-APC/C and is degraded by the pool of Cdc20 that avoids inhibition by the SAC.


Subject(s)
Cell Cycle Proteins/physiology , Kinesins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Ubiquitin-Protein Ligase Complexes/physiology , Anaphase-Promoting Complex-Cyclosome , Apc8 Subunit, Anaphase-Promoting Complex-Cyclosome , Cdc20 Proteins , Cell Cycle Proteins/metabolism , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/physiology , NIMA-Related Kinases , Prometaphase/physiology , Protein Binding , Protein Multimerization , Time Factors , Tumor Cells, Cultured , Ubiquitin-Protein Ligase Complexes/metabolism
10.
J Biol Chem ; 287(30): 24905-15, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22689581

ABSTRACT

Src family tyrosine kinases (SFKs) participate in mitotic signal transduction events, including mitotic entry, cleavage furrow ingression, and cytokinesis abscission. Although SFKs have been shown to associate with the mitotic spindle, the role of SFKs in mitotic spindle formation remains unclear. Here, we show that c-Src promotes proper spindle orientation in early prometaphase. Src localizes close to spindle poles in a manner independent of Src kinase activity. Three-dimensional analyses showed that Src inhibition induced spindle misorientation, exhibiting a tilting spindle in early prometaphase. Spindle misorientation is frequently seen in SYF cells, which harbor triple knock-out mutations of c-Src, c-Yes, and Fyn, and reintroduction of c-Src but not Fyn into SYF cells rescued spindle misorientation. Spindle misorientation was also observed upon Src inhibition under conditions in which Aurora B was inhibited. Inducible expression of c-Src promoted a properly oriented bipolar spindle, which was suppressed by Src inhibition. Aster formation was severely inhibited in SYF cells upon Aurora B inhibition, which was rescued by reintroduction of c-Src into SYF cells. Furthermore, reintroduction of c-Src facilitated microtubule regrowth from cold-induced depolymerization and accelerated M phase progression. These results suggest that c-Src is involved in spindle orientation through centrosome-mediated aster formation.


Subject(s)
Prometaphase/physiology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Spindle Apparatus/metabolism , Aurora Kinase B , Aurora Kinases , CSK Tyrosine-Protein Kinase , HeLa Cells , Humans , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-fyn/genetics , Spindle Apparatus/genetics , src-Family Kinases
11.
Development ; 139(11): 1947-55, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22513370

ABSTRACT

Homologous chromosome segregation errors during meiosis I are common and generate aneuploid embryos. Here, we provide a reason for this susceptibility to mis-segregation by live cell imaging of mouse oocytes. Our results show that stable kinetochore-microtubule attachments form in mid-prometaphase, 3-4 hours before anaphase. This coincided with the loss of Mad2 from kinetochores and with the start of anaphase-promoting complex/cyclosome (APC/C)-mediated cyclin B1 destruction. Therefore, the spindle assembly checkpoint (SAC) ceased to inhibit the APC/C from mid-prometaphase. This timing did not coincide with bivalent congression in one-third of all oocytes examined. Non-aligned bivalents were weakly positive for Mad2, under less tension than congressed bivalents and, by live-cell imaging, appeared to be in the process of establishing correct bi-orientation. The time from when the APC/C became active until anaphase onset was affected by the rate of loss of CDK1 activity, rather than by these non-aligned bivalents, which occasionally persisted until anaphase, resulting in homolog non-disjunction. We conclude that, in oocytes, a few erroneous attachments of bivalent kinetochores to microtubules do not generate a sufficient SAC 'wait anaphase' signal to inhibit the APC/C.


Subject(s)
Chromosome Segregation/physiology , Kinetochores/metabolism , Microtubules/metabolism , Oocytes/physiology , Prometaphase/physiology , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Cycle Proteins/metabolism , Cyclin B1/metabolism , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Immunoblotting , Mad2 Proteins , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Oocytes/metabolism , RNA, Complementary/genetics , Time Factors
12.
Cell Cycle ; 9(19): 3956-64, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20948288

ABSTRACT

The mechanisms that control E2F-1 activity are complex. We previously showed that Chk1 and Chk2 are required for E2F1 stabilization and p73 target gene induction following DNA damage. To gain further insight into the processes regulating E2F1 protein stability, we focused our investigation on the mechanisms responsible for regulating E2F1 turnover. Here we show that E2F1 is a substrate of the anaphase promoting complex or cyclosome (APC/C), a ubiquitin ligase that plays an important role in cell cycle progression. Ectopic expression of the APC/C activators Cdh1 and Cdc20 reduced the levels of co-expressed E2F-1 protein. Co-expression of DP1 with E2F1 blocked APC/C-induced E2F1 degradation, suggesting that the E2F1/DP1 heterodimer is protected from APC/C regulation. Following Cdc20 knockdown, E2F1 levels increased and remained stable in extracts over a time course, indicating that APC/C(Cdc20) is a primary regulator of E2F1 stability in vivo. Moreover, cell synchronization experiments showed that siRNA directed against Cdc20 induced an accumulation of E2F1 protein in prometaphase cells. These data suggest that APC/C(Cdc20) specifically targets E2F1 for degradation in early mitosis and reveal a novel mechanism for limiting free E2F1 levels in cells, failure of which may compromise cell survival and/or homeostasis.


Subject(s)
Cell Cycle Proteins/metabolism , E2F1 Transcription Factor/metabolism , Prometaphase/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Cdc20 Proteins , Cell Cycle Proteins/genetics , Checkpoint Kinase 1 , Checkpoint Kinase 2 , E2F1 Transcription Factor/genetics , HeLa Cells , Humans , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factor DP1/genetics , Transcription Factor DP1/metabolism , Ubiquitin-Protein Ligase Complexes/genetics
13.
Curr Biol ; 20(18): 1666-71, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20832310

ABSTRACT

The mitotic checkpoint maintains genomic stability by blocking the metaphase-anaphase transition until all kinetochores attach to spindle microtubules [1, 2]. However, some defects are not detected by this checkpoint. With low concentrations of microtubule-targeting agents, the checkpoint eventually becomes satisfied, though the spindles may be short and/or multipolar [3, 4] and the fidelity of chromosome distribution and cleavage completion are compromised. In real life, environmental toxins, radiation, or chemotherapeutic agents may lead to completed but inaccurate mitoses. It has been assumed that once the checkpoint is satisfied and cells divide, the daughter cells would proliferate regardless of prometaphase duration. However, when continuously exposed to microtubule inhibitors, untransformed cells eventually slip out of mitosis after 12-48 hr and arrest in G1 [5-8] (see also [9]). Interestingly, transient but prolonged treatments with nocodazole allow completion of mitosis, but the daughter cells arrest in interphase [10, 11] (see also [9, 12]). Here we characterize the relationship between prometaphase duration and the proliferative capacity of daughter cells. Our results reveal the existence of a mechanism that senses prometaphase duration; if prometaphase lasts >1.5 hr, this mechanism triggers a durable p38- and p53-dependent G1 arrest of the daughter cells despite normal division of their mothers.


Subject(s)
Cell Proliferation , Mitosis/physiology , Prometaphase/physiology , Antineoplastic Agents/metabolism , Cell Line , Humans , Leupeptins/metabolism , Nocodazole/metabolism
14.
J Cell Biol ; 190(4): 587-602, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20733055

ABSTRACT

The ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C) is activated at prometaphase by mitotic phosphorylation and binding of its activator, Cdc20. This initiates cyclin A degradation, whereas cyclin B1 is stabilized by the spindle checkpoint. Upon checkpoint release, the RXXL destruction box (D box) was proposed to direct cyclin B1 to core APC/C or Cdc20. In this study, we report that endogenous cyclin B1-Cdk1 is recruited to checkpoint-inhibited, phosphorylated APC/C in prometaphase independently of Cdc20 or the cyclin B1 D box. Like cyclin A, cyclin B1 binds the APC/C by the Cdk cofactor Cks and the APC3 subunit. Prior binding to APC/C(Cdc20) makes cyclin B1 a better APC/C substrate in metaphase, driving mitotic exit and cytokinesis. We conclude that in prometaphase, the phosphorylated APC/C can recruit both cyclin A and cyclin B1 in a Cks-dependent manner. This suggests that the spindle checkpoint blocks D box recognition of APC/C-bound cyclin B1, whereas distinctive complexes between the N terminus of cyclin A and Cdc20 evade checkpoint control.


Subject(s)
CDC2 Protein Kinase/metabolism , Carrier Proteins/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinases/metabolism , Mitosis/physiology , Prometaphase/physiology , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , CDC2 Protein Kinase/genetics , CDC2-CDC28 Kinases , Carrier Proteins/genetics , Cdc20 Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cyclin A/genetics , Cyclin A/metabolism , Cyclin B1/genetics , Cyclin-Dependent Kinases/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nocodazole/metabolism , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/metabolism , Purines/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Roscovitine , Securin , Tubulin Modulators/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligase Complexes/genetics
15.
Postepy Biochem ; 56(1): 29-40, 2010.
Article in Polish | MEDLINE | ID: mdl-20499679

ABSTRACT

During cell division the bipolar microtubular mitotic spindle ensures faithful segregation of daughter chromosomes and appearance of the cytokinetic membrane in an equatorial area separating genetically identical daughter cells. This process proceeds in consecutive morphological stages of prophase, metaphase, anaphase and cytokinesis. The progress in embryology and oncology concerns the new data about intervening mechanisms of rotation of a bipolar spindle in prophase and the change of the position of a mitotic spindle in anaphase that result in an asymmetric and differential mitoses. The aim of this review is a discussion of some of molecular and signaling mechanisms which regulate position of mitotic spindles in different types of cells. It turns out that the knowledge of receptor-dependent and receptor-independent molecular mechanisms controlling geometry and localization of cytokinesis in some human cells and in early stages of development of C. elegans opens the new important research fields.


Subject(s)
Cytokinesis/physiology , Mitosis/physiology , Anaphase/physiology , Animals , Caenorhabditis elegans/growth & development , Cell Cycle/physiology , Fibronectins/metabolism , Humans , Metaphase/physiology , Prometaphase/physiology , Signal Transduction/physiology
16.
J Cell Physiol ; 223(2): 327-34, 2010 May.
Article in English | MEDLINE | ID: mdl-20054826

ABSTRACT

Cdc20, an activator of the anaphase promoting complex/cyclosome (APC/C) ubiquitin ligase, initiates the destruction of key mitotic regulators to facilitate mitosis, while it is negatively regulated by the spindle assembly checkpoint (SAC) to prevent premature anaphase entry. Activation of the p38 mitogen-activated protein kinase could contribute to mitotic arrest, but the underlying mechanism is unknown. Here we report a novel pathway in which the p38 signaling triggers Cdc20 destruction under SAC elicited by cadmium, a human carcinogen. We found that the cadmium-induced prometaphase arrest was linked to decreased Cdc20 and accumulated cyclin A protein levels in human cells, whereas the activity of cyclin B1-Cdk1 was unaffected. The Cdc20 half-life was markedly shortened along with its ubiquitination and degradation via 26S proteasome in cadmium-treated asynchronous or G(2)-enriched cells. Depletion of APC3 markedly suppressed the cadmium-induced Cdc20 ubiquitination and proteolysis, while depletion of Cdh1, another activator of APC/C, did not. Intriguingly, blockage of p38 activity restored the Cdc20 levels for continuing mitosis under cadmium, while inhibition of JNK activity had no effect. The cadmium-induced Cdc20 proteolysis was also suppressed during transient depletion of p38alpha or stable expression a dominant negative form of p38. Inhibition of p38 abolished the induction of Mad2-Cdc20-APC3 complex by cadmium. Moreover, forced expression of MKK6-p38 signaling could promote Cdc20 degradation in a Cdh1-independent APC/C pathway. In summary, accelerated ubiquitination and proteolysis of Cdc20 is essential for prometaphase arrest that is mediated via the p38 signaling during SAC activation.


Subject(s)
Cell Cycle Proteins/metabolism , Genes, cdc/physiology , Spindle Apparatus/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Anaphase-Promoting Complex-Cyclosome , Antigens, CD , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome , Cadherins/metabolism , Cadmium/pharmacology , Cdc20 Proteins , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cyclin A/drug effects , Cyclin A/metabolism , Enzyme Inhibitors/pharmacology , Genes, cdc/drug effects , Humans , MAP Kinase Kinase 6/drug effects , MAP Kinase Kinase 6/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogens/pharmacology , Peptide Hydrolases/drug effects , Peptide Hydrolases/metabolism , Prometaphase/drug effects , Prometaphase/physiology , Spindle Apparatus/drug effects , Ubiquitination/drug effects , Ubiquitination/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
17.
Science ; 326(5955): 991-4, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19965510

ABSTRACT

Two critical stages of mammalian oocyte regulation are prophase I arrest, which is important for sustaining the oocyte pool, and the progression through meiosis I (MI) to produce fertilizable eggs. We have found that the spindle assembly checkpoint protein BubR1 regulates both stages in mouse oocytes. We show that oocytes depleted of BubR1 cannot sustain prophase I arrest and readily undergo germinal vesicle breakdown, a marker for reentry into MI. BubR1-depleted oocytes then arrest before completing MI, marked by failure of polar body extrusion. Both meiotic defects in BubR1-depleted oocytes are due to reduced activity of the master regulator known as the anaphase-promoting complex (APC), brought about through diminished levels of the APC coactivator Cdh1.


Subject(s)
Meiosis/physiology , Meiotic Prophase I/physiology , Oocytes/physiology , Prometaphase/physiology , Prometaphase/radiation effects , Protein Serine-Threonine Kinases/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Carrier Proteins/metabolism , Cdc20 Proteins , Cdh1 Proteins , Cell Cycle Proteins/metabolism , Cyclin B1/metabolism , Female , Gene Silencing , Mice , Protein Serine-Threonine Kinases/genetics , Securin , Ubiquitin-Protein Ligase Complexes/metabolism
18.
Cancer Res ; 69(6): 2314-23, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276349

ABSTRACT

The Aurora (Ipl) kinase family plays important roles in the regulation of mitosis and tumorigenesis. The tumor suppressor RASSF1A controls mitotic progression by regulating anaphase-promoting complex (APC)-Cdc20 activity and microtubule stability, but the mechanism by which this action is regulated has not been previously established. Here, we show that Aurora A and B associate with and phosphorylate RASSF1A on serine 203 in vivo at different times and in different subcellular compartments during mitosis. Notably, both depletion of Aurora A by RNA interference and expression of a nonphosphorylatable RASSF1A (S203A) mutant gene led to a marked delay in prometaphase progression. This is likely because of the failure of RASSF1A to dissociate from Cdc20, constitutive inhibition of APC-Cdc20, and accumulation of mitotic cyclins. In contrast, the delay in prometaphase progression caused by Aurora A depletion was largely normalized by phosphomimetic RASSF1A (S203D). Finally, RASSF1A phosphorylation on serine 203 was up-regulated in Aurora A-overexpressing human tumors. These findings indicate that Aurora A plays a critical role in RASSF1A-APC-Cdc20 regulatory mechanisms that control normal prometaphase progression and that are involved in tumorigenesis. [Cancer Res 2009;69(6):2314-23.


Subject(s)
Cell Cycle Proteins/metabolism , Prometaphase/physiology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Aurora Kinases , Cdc20 Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Division/physiology , G2 Phase/physiology , HeLa Cells , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mitosis/physiology , Mutagenesis, Site-Directed , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
19.
J Virol ; 83(4): 1700-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19052088

ABSTRACT

We previously observed that high-risk human papillomavirus type 16 (HPV16) E7 expression leads to the delocalization of dynein from mitotic spindles (C. L. Nguyen, M. E. McLaughlin-Drubin, and K. Munger, Cancer Res. 68:8715-8722, 2008). Here, we show that HPV16 E7 associates with nuclear mitotic apparatus protein 1 (NuMA) and that NuMA binding and the ability to induce dynein delocalization map to similar carboxyl-terminal sequences of E7. Additionally, we show that the delocalization of dynein from mitotic spindles by HPV16 E7 and the interaction between HPV16 E7 and NuMA correlate with the induction of defects in chromosome alignment during prometaphase even in cells with normal centrosome numbers. Furthermore, low-risk HPV6b and HPV11 E7s also associate with NuMA and also induce a similar mitotic defect. It is possible that the disruption of mitotic events by HPV E7, via targeting of the NuMA/dynein complex and potentially other NuMA-containing complexes, contributes to viral maintenance and propagation potentially through abrogating the differentiation program of the infected epithelium. Furthermore, in concert with activities specific to high-risk HPV E6 and E7, such as the inactivation of the p53 and pRB tumor suppressors, respectively, the disruption of the NuMA/dynein network may result in mitotic errors that would make an infected cell more prone to the accumulation of aneuploidy even in the absence of supernumerary centrosomes.


Subject(s)
Antigens, Nuclear/metabolism , Human papillomavirus 16/pathogenicity , Mitosis , Nuclear Matrix-Associated Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Cell Cycle Proteins , Cell Line , Chromosomes/metabolism , Dyneins/metabolism , Humans , Papillomavirus E7 Proteins , Prometaphase/physiology , Protein Binding , Protein Interaction Mapping
20.
Biochim Biophys Acta ; 1786(1): 24-31, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18472014

ABSTRACT

Aneuploidy, an abnormal number of chromosomes, is a trait shared by most solid tumors. Chromosomal instability (CIN) manifested as aneuploidy might promote tumorigenesis and cause increased resistance to anti-cancer therapies. The mitotic checkpoint or spindle assembly checkpoint is a major signaling pathway involved in the prevention of CIN. We review current knowledge on the contribution of misregulation of mitotic checkpoint proteins to tumor formation and will address to what extent this contribution is due to chromosome segregation errors directly. We propose that both checkpoint and non-checkpoint functions of these proteins contribute to the wide array of oncogenic phenotypes seen upon their misregulation.


Subject(s)
Aneuploidy , Mitosis/physiology , Neoplasms/etiology , Animals , Cdc20 Proteins , Cell Cycle Proteins/physiology , Cell Survival , Chromosomal Instability , Chromosome Segregation/physiology , Humans , Mad2 Proteins , Mice , Models, Animal , Nuclear Proteins/physiology , Prometaphase/physiology , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...