scispace - formally typeset
Search or ask a question

Showing papers on "Metaphase published in 2009"


Journal ArticleDOI
TL;DR: Apart from certain minor but well defined chromosome regions with especially strong fluorescence, which are subject to certain individual variations, the fluorescence patterns were shown to be quite stable and reproducible.
Abstract: Certain fluorescent DNA-binding compounds, among them quinacrine mustard and quinacrine, give characteristic banding patterns in human metaphase chromosomes; these patterns can be used to identify all the 24 chromosome types as well as chromosome aberrations. The patterns given by quinacrine mustard are especially clear and stable and are suitable for chromosome identification either visually—preferably after contrast enhancement by photography—or by photometric methods. The typical fluorescence pattern of each chromosome type is described. The reproducibility and variability of the patterns have been analysed by photometric measurements of the patterns in a material of about 5000 chromosomes from 14 healthy subjects. Apart from certain minor but well defined chromosome regions with especially strong fluorescence, which are subject to certain individual variations, the fluorescence patterns were shown to be quite stable and reproducible.

623 citations


Journal ArticleDOI
02 Oct 2009-Cell
TL;DR: These findings support a model in which CSR-1 complexes target protein-coding domains to promote their proper organization within the holocentric chromosomes of C. elegans.

431 citations


Journal ArticleDOI
10 Aug 2009-PLOS ONE
TL;DR: The data suggest a model by which merotelic kinetochore attachments can easily be established in multipolar prometaphases, and a spindle pole coalescence mechanism as a major contributor to chromosome instability in cancer cells.
Abstract: Many cancer cells display a CIN (Chromosome Instability) phenotype, by which they exhibit high rates of chromosome loss or gain at each cell cycle. Over the years, a number of different mechanisms, including mitotic spindle multipolarity, cytokinesis failure, and merotelic kinetochore orientation, have been proposed as causes of CIN. However, a comprehensive theory of how CIN is perpetuated is still lacking. We used CIN colorectal cancer cells as a model system to investigate the possible cellular mechanism(s) underlying CIN. We found that CIN cells frequently assembled multipolar spindles in early mitosis. However, multipolar anaphase cells were very rare, and live-cell experiments showed that almost all CIN cells divided in a bipolar fashion. Moreover, fixed-cell analysis showed high frequencies of merotelically attached lagging chromosomes in bipolar anaphase CIN cells, and higher frequencies of merotelic attachments in multipolar vs. bipolar prometaphases. Finally, we found that multipolar CIN prometaphases typically possessed γ-tubulin at all spindle poles, and that a significant fraction of bipolar metaphase/early anaphase CIN cells possessed more than one centrosome at a single spindle pole. Taken together, our data suggest a model by which merotelic kinetochore attachments can easily be established in multipolar prometaphases. Most of these multipolar prometaphase cells would then bi-polarize before anaphase onset, and the residual merotelic attachments would produce chromosome mis-segregation due to anaphase lagging chromosomes. We propose this spindle pole coalescence mechanism as a major contributor to chromosome instability in cancer cells.

421 citations


Journal ArticleDOI
15 May 2009-Cell
TL;DR: Delta analysis revealed that compliant linkages are restricted to the proximity of chromatin, suggested a model for how the KMN (KNL1/Mis12 complex/Ndc80 complex) network provides microtubule attachment and generates pulling forces from depolymerization, and identified an intrakinetochore molecular switch that may function in controlling checkpoint activity.

351 citations


Journal ArticleDOI
TL;DR: The geometry of the core structure of the budding yeast Kinetochore reported here is remarkably similar to that found in mammalian kinetochores, indicating that kinetchore structure is conserved in eukaryotes with either point or regional centromeres.

200 citations


Journal ArticleDOI
TL;DR: It is suggested that this network plays a major role in silencing the spindle checkpoint when chromosomes are aligned at metaphase to allow timely anaphase onset and mitotic exit.

174 citations


Journal ArticleDOI
TL;DR: The data support the idea that congression allows unattached chromosomes to move to the middle of the spindle where they have a higher probability of establishing connections with both spindle poles, and are also used to maintain stable chromosome alignment at theSpindle equator.
Abstract: Proper chromosome congression (the process of aligning chromosomes on the spindle) contributes to accurate and faithful chromosome segregation. It is widely accepted that congression requires kinetochore fibres (K-fibres), microtubule bundles that extend from the kinetochores to spindle poles. Here, we demonstrate that chromosomes in human cells co-depleted of HSET (human kinesin-14) and hNuf2 (human Ndc80/Hec1-complex component) can congress to the metaphase plate in the absence of K-fibres. However, the chromosomes are not stably maintained at the metaphase plate under these conditions. Chromosome congression in HSET + hNuf2 co-depleted cells required the plus-end directed motor CENP-E (centromere protein E; kinesin-7 family member), which has been implicated in the gliding of mono-oriented kinetochores alongside adjacent K-fibres. Thus, proper end-on attachment of kinetochores to microtubules is not necessary for chromosome congression. Instead, our data support the idea that congression allows unattached chromosomes to move to the middle of the spindle where they have a higher probability of establishing connections with both spindle poles. These bi-oriented connections are also used to maintain stable chromosome alignment at the spindle equator.

148 citations


Journal ArticleDOI
TL;DR: It is demonstrated that CI disrupts earlier interphase cell cycle events in the male pronucleus and proposed that these CI-induced interphase defects in de novo nucleosome assembly and replication are the cause of the observed mitotic condensation and segregation defects.
Abstract: Wolbachia is a bacteria endosymbiont that rapidly infects insect populations through a mechanism known as cytoplasmic incompatibility (CI). In CI, crosses between Wolbachia-infected males and uninfected females produce severe cell cycle defects in the male pronucleus resulting in early embryonic lethality. In contrast, viable progeny are produced when both parents are infected (the Rescue cross). An important consequence of CI–Rescue is that infected females have a selective advantage over uninfected females facilitating the rapid spread of Wolbachia through insect populations. CI disrupts a number of prophase and metaphase events in the male pronucleus, including Cdk1 activation, chromosome condensation, and segregation. Here, we demonstrate that CI disrupts earlier interphase cell cycle events. Specifically, CI delays the H3.3 and H4 deposition that occurs immediately after protamine removal from the male pronucleus. In addition, we find prolonged retention of the replication factor PCNA in the male pronucleus into metaphase, indicating progression into mitosis with incompletely replicated DNA. We propose that these CI-induced interphase defects in de novo nucleosome assembly and replication are the cause of the observed mitotic condensation and segregation defects. In addition, these interphase chromosome defects likely activate S-phase checkpoints, accounting for the previously described delays in Cdk1 activation. These results have important implications for the mechanism of Rescue and other Wolbachia-induced phenotypes.

145 citations


Journal ArticleDOI
TL;DR: It is proposed that ensheathment by lateral microtubule bundles places spatial constraints on the chromosomes, thereby promoting biorientation, and that localized motors mediate movement along these bundles, thereby promote alignment.
Abstract: Although centrosomes serve to organize microtubules in most cell types, oocyte spindles form and mediate meiotic chromosome segregation in their absence. Here, we used high-resolution imaging of both bipolar and experimentally generated monopolar spindles in Caenorhabditis elegans to reveal a surprising organization of microtubules and chromosomes within acentrosomal structures. We found that homologous chromosome pairs (bivalents) are surrounded by microtubule bundles running along their sides, whereas microtubule density is extremely low at chromosome ends despite a high concentration of kinetochore proteins at those regions. Furthermore, we found that the chromokinesin KLP-19 (kinesin-like protein 19) is targeted to a ring around the centre of each bivalent and provides a polar ejection force that is required for congression. Together, these observations create a new picture of chromosome-microtubule association in acentrosomal spindles and reveal a mechanism by which metaphase alignment can be achieved using this organization. Specifically, we propose that ensheathment by lateral microtubule bundles places spatial constraints on the chromosomes, thereby promoting biorientation, and that localized motors mediate movement along these bundles, thereby promoting alignment.

144 citations


Journal ArticleDOI
TL;DR: It is shown that condensin ATPase activity is an important regulator of centromere stiffness and function and is dispensable for the normal level of compaction of centromeres, which probably depends on other factors that control higher-order chromatin folding.
Abstract: When chromosomes are aligned and bioriented at metaphase, the elastic stretch of centromeric chromatin opposes pulling forces exerted on sister kinetochores by the mitotic spindle. Here we show that condensin ATPase activity is an important regulator of centromere stiffness and function. Condensin depletion decreases the stiffness of centromeric chromatin by 50% when pulling forces are applied to kinetochores. However, condensin is dispensable for the normal level of compaction (rest length) of centromeres, which probably depends on other factors that control higher-order chromatin folding. Kinetochores also do not require condensin for their structure or motility. Loss of stiffness caused by condensin-depletion produces abnormal uncoordinated sister kinetochore movements, leads to an increase in Mad2(+) kinetochores near the metaphase plate and delays anaphase onset.

144 citations


Journal ArticleDOI
TL;DR: This work quantified the relationship between spindle elongation and cell size in the early embryo of Caenorhabditis elegans and proposed possible models for cell-size-dependent and Galpha-dependent modes of elongation.

Journal ArticleDOI
Y. Manolova1, George Manolov1, J. Kieler1, Albert Levan1, George Klein1 
TL;DR: An improved resolution of the G-band pattern has been achieved in prosome analysis of the (8;14) translocation of Burkitt’s lymphoma and the concept of local prosomisation was introduced and discussed.
Abstract: By means of the ”prosome” analysis technique. by which chromosomes are studied at early metaphase and even at prophase. an improved resolution of the G-band pattern has been achieved. The 2 chromosome regions of especial significance in the (8;14) translocation of Burkitt’s lymphoma, 8q24 and 14q32, which in the Pans karyotype were represented as uniformly negative, in prosome analysis resolved into 5 and 6 subbands respectively. The (8;14) translocation was shown to be reciprocal and the breaking points could be established in both chromosomes with more precision than earlier. It was noticed that the segment of No. 8. translocated onto No. 14, exhibited somewhat different morphologic features in translocated than in normal position. The concept of local prosomisation was introduced and discussed. Yanka Manolova, Oncological Research Institute, Sofia 1156, Bulgaria

Journal ArticleDOI
TL;DR: In this paper, an R2R3-type transcription factor gene, AtMYB59, was found to be involved in the regulation of cell cycle progression and root growth.
Abstract: MYB proteins play important roles in eukaryotic organisms. In plants, the R1R2R3-type MYB proteins function in cell cycle control. However, whether the R2R3-type MYB protein is also involved in the cell division process remains unknown. Here, we report that an R2R3-type transcription factor gene, AtMYB59, is involved in the regulation of cell cycle progression and root growth. The AtMYB59 protein is localized in the nuclei of onion epidermal cells and has transactivation activity. Expression of AtMYB59 in yeast cells suppresses cell proliferation, and the transformants have more nuclei and higher aneuploid DNA content with longer cells. Mutation in the conserved domain of AtMYB59 abolishes its effects on yeast cell growth. In synchronized Arabidopsis cell suspensions, the AtMYB59 gene is specifically expressed in the S phase during cell cycle progression. Expression and promoter-GUS analysis reveals that the AtMYB59 gene is abundantly expressed in roots. Transgenic plants overexpressing AtMYB59 have shorter roots compared with wild-type plants (Arabidopsis accession Col-0), and around half of the mitotic cells in root tips are at metaphase. Conversely, the null mutant myb59-1 has longer roots and fewer mitotic cells at metaphase than Col, suggesting that AtMYB59 may inhibit root growth by extending the metaphase of mitotic cells. AtMYB59 regulates many downstream genes, including the CYCB1;1 gene, probably through binding to MYB-responsive elements. These results support a role for AtMYB59 in cell cycle regulation and plant root growth.

Journal ArticleDOI
TL;DR: It is revealed that, after NEB, KLP61F drives persistent MT bundling and the outward sliding of antiparallel MTs, thereby contributing to several processes that all appear insensitive to cortical disruption.
Abstract: We used antibody microinjection and genetic manipulations to dissect the various roles of the homotetrameric kinesin-5, KLP61F, in astral, centrosome-controlled Drosophila embryo spindles and to test the hypothesis that it slides apart interpolar (ip) microtubules (MT), thereby controlling poleward flux and spindle length. In wild-type and Ncd null mutant embryos, anti-KLP61F dissociated the motor from spindles, producing a spatial gradient in the KLP61F content of different spindles, which was visible in KLP61F-GFP transgenic embryos. The resulting mitotic defects, supported by gene dosage experiments and time-lapse microscopy of living klp61f mutants, reveal that, after NEB, KLP61F drives persistent MT bundling and the outward sliding of antiparallel MTs, thereby contributing to several processes that all appear insensitive to cortical disruption. KLP61F activity contributes to the poleward flux of both ipMTs and kinetochore MTs and to the length of the metaphase spindle. KLP61F activity maintains the prometaphase spindle by antagonizing Ncd and another unknown force-generator and drives anaphase B, although the rate of spindle elongation is relatively insensitive to the motor's concentration. Finally, KLP61F activity contributes to normal chromosome congression, kinetochore spacing, and anaphase A rates. Thus, a KLP61F-driven sliding filament mechanism contributes to multiple aspects of mitosis in this system.

Journal ArticleDOI
TL;DR: The predominant role of PBD-dependent binding on smooth chromosome congression at metaphase is demonstrated and the high-throughput screening system developed shows it to be essential for Plk1 localization and substrate targeting.

Journal ArticleDOI
TL;DR: It is shown that NDR 1 is required for accurate chromosome alignment at metaphase in HeLa cells; depletion of NDR1, Fry, or MST2 caused mitotic chromosome misalignment.

Journal ArticleDOI
TL;DR: Most of the double minute chromosomes, in spite of their lack of detectable centromeric activity, were included into the telophase nuclei, and some micronuclei were found, however, probably leading to the loss of a proportion of the DMs.
Abstract: Normally, double minute (DM) chromosomes are extremely difficult to observe at other mitotic stages than prophase and metaphase. In a certain clonal subline of the mouse ascites tumor SEWA, however, the DMs were unusually distinct and could be followed through all mitotic stages. Their behavior was extraordinary: During metaphase-anaphase they exhibited no response whatever towards the spindle forces, and it was concluded that the DMs were without functioning centromere. Instead, at metaphase they were attached to or enclosed by the nucleolar matter persisting around the chromosome ends, and at anaphase they were transported to the poles together with the nucleolar matter, sticking to the daughter chromosomes. The daughter halves of each DM were usually carried to the same pole. Thus most of the DMs, in spite of their lack of detectable centromeric activity, were included into the telophase nuclei. Some micronuclei were found, however, probably leading to the loss of a proportion of the DMs. The content of DMs in a certain tumor cell population therefore must depend on the balance between this loss and the gain due to a positive selection value of cells containing DMs.

Journal ArticleDOI
TL;DR: It is proposed that a low stringency of the pachytene checkpoint could help to increase the chances that spermatocytes with synaptic defects will complete meiotic divisions and differentiate into viable gametes, explaining the multitude of natural Robertsonian populations described in the mouse.
Abstract: Meiosis is a complex type of cell division that involves homologous chromosome pairing, synapsis, recombination, and segregation. When any of these processes is altered, cellular checkpoints arrest meiosis progression and induce cell elimination. Meiotic impairment is particularly frequent in organisms bearing chromosomal translocations. When chromosomal translocations appear in heterozygosis, the chromosomes involved may not correctly complete synapsis, recombination, and/or segregation, thus promoting the activation of checkpoints that lead to the death of the meiocytes. In mammals and other organisms, the unsynapsed chromosomal regions are subject to a process called meiotic silencing of unsynapsed chromatin (MSUC). Different degrees of asynapsis could contribute to disturb the normal loading of MSUC proteins, interfering with autosome and sex chromosome gene expression and triggering a massive pachytene cell death. We report that in mice that are heterozygous for eight multiple simple Robertsonian translocations, most pachytene spermatocytes bear trivalents with unsynapsed regions that incorporate, in a stage-dependent manner, proteins involved in MSUC (e.g., γH2AX, ATR, ubiquitinated-H2A, SUMO-1, and XMR). These spermatocytes have a correct MSUC response and are not eliminated during pachytene and most of them proceed into diplotene. However, we found a high incidence of apoptotic spermatocytes at the metaphase stage. These results suggest that in Robertsonian heterozygous mice synapsis defects on most pachytene cells do not trigger a prophase-I checkpoint. Instead, meiotic impairment seems to mainly rely on the action of a checkpoint acting at the metaphase stage. We propose that a low stringency of the pachytene checkpoint could help to increase the chances that spermatocytes with synaptic defects will complete meiotic divisions and differentiate into viable gametes. This scenario, despite a reduction of fertility, allows the spreading of Robertsonian translocations, explaining the multitude of natural Robertsonian populations described in the mouse.

Journal ArticleDOI
TL;DR: It is shown that the analysis of protein localisation is useful to refine a phenotypic profile and identified C13orf3 (Ska3) as a new component of the Ska complex and show a direct interaction with a regulatory subunit of the protein phosphatase PP2A.
Abstract: Proliferation of mammalian cells requires the coordinated function of many proteins to accurately divide a cell into two daughter cells. Several RNAi screens have identified previously uncharacterised genes that are implicated in mammalian cell division. The molecular function for these genes needs to be investigated to place them into pathways. Phenotypic profiling is a useful method to assign putative functions to uncharacterised genes. Here, we show that the analysis of protein localisation is useful to refine a phenotypic profile. We show the utility of this approach by defining a function of the previously uncharacterised gene C13orf3 during cell division. C13orf3 localises to centrosomes, the mitotic spindle, kinetochores, spindle midzone, and the cleavage furrow during cell division and is specifically phosphorylated during mitosis. Furthermore, C13orf3 is required for centrosome integrity and anaphase onset. Depletion by RNAi leads to mitotic arrest in metaphase with an activation of the spindle assembly checkpoint and loss of sister chromatid cohesion. Proteomic analyses identify C13orf3 (Ska3) as a new component of the Ska complex and show a direct interaction with a regulatory subunit of the protein phosphatase PP2A. All together, these data identify C13orf3 as an important factor for metaphase to anaphase progression and highlight the potential of combined RNAi screening and protein localisation analyses.

Journal ArticleDOI
TL;DR: It is shown that tension uniformity at metaphase Kinetochores and subsequent anaphase synchrony in Drosophila S2 cells are promoted by spindle microtubule flux, and proposed that flux-dependent equalization of kinetochore tensions ensures a timely and uniform maturation of kinETochore–microtubule interfaces necessary for error-free and coordinated segregation of chromosomes inAnaphase.
Abstract: The synchronous movement of chromosomes during anaphase ensures their correct inheritance in every cell division. This reflects the uniformity of spindle forces acting on chromosomes and their simultaneous entry into anaphase. Although anaphase onset is controlled by the spindle assembly checkpoint, it remains unknown how spindle forces are uniformly distributed among different chromosomes. In this paper, we show that tension uniformity at metaphase kinetochores and subsequent anaphase synchrony in Drosophila S2 cells are promoted by spindle microtubule flux. These results can be explained by a mechanical model of the spindle where microtubule poleward translocation events associated with flux reflect relaxation of the kinetochore–microtubule interface, which accounts for the redistribution and convergence of kinetochore tensions in a timescale comparable to typical metaphase duration. As predicted by the model, experimental acceleration of mitosis precludes tension equalization and anaphase synchrony. We propose that flux-dependent equalization of kinetochore tensions ensures a timely and uniform maturation of kinetochore–microtubule interfaces necessary for error-free and coordinated segregation of chromosomes in anaphase.

Journal ArticleDOI
TL;DR: The prometaphase movement of achiasmate chromosomes is a congression-like process in which the two homologs undergo both separation and rejoining events that result in the either loss or establishment of proper kinetochore co-orientation.
Abstract: In Drosophila oocytes achiasmate homologs are faithfully segregated to opposite poles at meiosis I via a process referred to as achiasmate homologous segregation. We observed that achiasmate homologs display dynamic movements on the meiotic spindle during mid-prometaphase. An analysis of living prometaphase oocytes revealed both the rejoining of achiasmate X chromosomes initially located on opposite half-spindles and the separation toward opposite poles of two X chromosomes that were initially located on the same half spindle. When the two achiasmate X chromosomes were positioned on opposite halves of the spindle their kinetochores appeared to display proper co-orientation. However, when both Xs were located on the same half spindle their kinetochores appeared to be oriented in the same direction. Thus, the prometaphase movement of achiasmate chromosomes is a congression-like process in which the two homologs undergo both separation and rejoining events that result in the either loss or establishment of proper kinetochore co-orientation. During this period of dynamic chromosome movement, the achiasmate homologs were connected by heterochromatic threads that can span large distances relative to the length of the developing spindle. Additionally, the passenger complex proteins Incenp and Aurora B appeared to localize to these heterochromatic threads. We propose that these threads assist in the rejoining of homologs and the congression of the migrating achiasmate homologs back to the main chromosomal mass prior to metaphase arrest.

Journal ArticleDOI
TL;DR: The results of this study further strengthen the hypothesis that Separase might be an oncogene, whose overexpression induces tumorigenesis, and indicates thatSeparase overeexpression and aberrant nuclear localization are common in many tumor types and may predict outcome in some human cancers.
Abstract: Purpose: Separase, an endopeptidase, plays a pivotal role in chromosomal segregation by separating sister chromatids during the metaphase to anaphase transition. Using a mouse mammary tumor model we have recently shown that overexpression of Separase induces aneuploidy and tumorigenesis (Zhang et al., Proc Natl Acad Sci 2008;105:13033). In the present study, we have investigated the expression level of Separase across a wide range of human tumors. Experimental Design: To examine the expression levels and localization of Separase in human tumors, we have performed immunofluorescence microscopy using human Separase antibody and tumor tissue arrays from osteosarcoma, colorectal, breast, and prostate cancers with appropriate normal controls. Results: We show that Separase is significantly overexpressed in osteosarcoma, breast, and prostate tumor specimens. There is a strong correlation of tumor status with the localization of Separase into the nucleus throughout all stages of the cell cycle. Unlike the normal control tissues, where Separase localization is exclusively cytoplasmic in nondividing cells, human tumor samples show significantly higher number of resting cells with a strong nuclear Separase staining. Additionally, overexpression of Separase transcript strongly correlates with high incidence of relapse, metastasis, and lower 5-year overall survival rate in breast and prostate cancer patients. Conclusion: These results further strengthen our hypothesis that Separase might be an oncogene, whose overexpression induces tumorigenesis, and indicates that Separase overexpression and aberrant nuclear localization are common in many tumor types and may predict outcome in some human cancers.

Journal ArticleDOI
Jingyan Fu1, Minglei Bian, Junjun Liu, Qing Jiang, Chuanmao Zhang 
TL;DR: It is proposed that the presence of Gly or Asn at a single site assigns Aurora-A and -B to their respective partners and thus to their distinctive subcellular localizations and functions.
Abstract: Aurora kinase-A and -B are key regulators of the cell cycle and tumorigenesis. It has remained a mystery why these 2 Aurora kinases, although highly similar in protein sequence and structure, are distinct in subcellular localization and function. Here, we report the striking finding that a single amino acid residue is responsible for these differences. We replaced the Gly-198 of Aurora-A with the equivalent residue Asn-142 of Aurora-B and found that in HeLa cells, Aurora-AG198N was recruited to the inner centromere in metaphase and the midzone in anaphase, reminiscent of the Aurora-B localization. Moreover, Aurora-AG198N compensated for the loss of Aurora-B in chromosome misalignment and cell premature exit from mitosis. Furthermore, Aurora-AG198N formed a complex with the Aurora-B partners, INCENP and Survivin, and its localization depended on this interaction. Aurora-AG198N phosphorylated the Aurora-B substrates INCENP and Survivin in vitro. Therefore, we propose that the presence of Gly or Asn at a single site assigns Aurora-A and -B to their respective partners and thus to their distinctive subcellular localizations and functions.

Journal ArticleDOI
TL;DR: It is concluded that a Cdk1 phosphorylated form of LIC1 is required to remove Mad1/2 and Zw10 but not BubR1 from kinetochores during spindle assembly checkpoint silencing.
Abstract: The spindle assembly checkpoint monitors microtubule attachment to kinetochores and tension across sister kinetochores to ensure accurate division of chromosomes between daughter cells. Cytoplasmic dynein functions in the checkpoint, apparently by moving critical checkpoint components off kinetochores. The dynein subunit required for this function is unknown. Here we show that human cells depleted of dynein light intermediate chain 1 (LIC1) delay in metaphase with increased interkinetochore distances; dynein remains intact, localised and functional. The checkpoint proteins Mad1/2 and Zw10 localise to kinetochores under full tension, whereas BubR1 is diminished at kinetochores. Metaphase delay and increased interkinetochore distances are suppressed by depletion of Mad1, Mad2 or BubR1 or by re-expression of wtLIC1 or a Cdk1 site phosphomimetic LIC1 mutant, but not Cdk1-phosphorylation-deficient LIC1. When the checkpoint is activated by microtubule depolymerisation, Mad1/2 and BubR1 localise to kinetochores. We conclude that a Cdk1 phosphorylated form of LIC1 is required to remove Mad1/2 and Zw10 but not BubR1 from kinetochores during spindle assembly checkpoint silencing.

Journal ArticleDOI
TL;DR: It is proposed that Aurora B/Ipl1-facilitated bi-orientation is stabilised in response to tension at Kinetochores by dephosphorylation of Dam1, resulting in termination of kinetochore-microtubule attachment turnover.
Abstract: The conserved Aurora B protein kinase (Ipl1 in Saccharomyces cerevisiae) is essential for ensuring that sister kinetochores become attached to microtubules from opposite spindle poles (bi-orientation) before anaphase onset. When sister chromatids become attached to microtubules from a single pole, Aurora B/Ipl1 facilitates turnover of kinetochore-microtubule attachments. This process requires phosphorylation by Aurora B/Ipl1 of kinetochore components such as Dam1 in yeast. Once bi-orientation is established and tension is applied on kinetochores, Aurora B/Ipl1 must stop promoting this turnover, otherwise correct attachment would never be stabilised. How this is achieved remains elusive: it might be due to dephosphorylation of Aurora B/Ipl1 substrates at kinetochores, or might take place independently, for example because of conformational changes in kinetochores. Here, we show that Ipl1-dependent phosphorylation at crucial sites on Dam1 is maximal during S phase and minimal during metaphase, matching the cell cycle window when chromosome bi-orientation occurs. Intriguingly, when we reduced tension at kinetochores through failure to establish sister chromatid cohesion, Dam1 phosphorylation persisted in metaphase-arrested cells. We propose that Aurora B/Ipl1-facilitated bi-orientation is stabilised in response to tension at kinetochores by dephosphorylation of Dam1, resulting in termination of kinetochore-microtubule attachment turnover.

Journal ArticleDOI
TL;DR: It is shown that absence of Ph1 causes increased transcription of Asynapsis 1 (ASY1), a gene that encodes an axial-element-associated protein that is essential for synapsis and cross-over formation in Arabidopsis and rice, and that Ph1 has a gene regulatory role, which is consistent with its suggested genetic identity as a Cdk-like gene.
Abstract: During meiosis, chromosomes are sorted into homologous pairs as a preface to their intimate association via recombination and synapsis. However, little is known about the mechanism used to distinguish homologous chromosomes from other chromosomes present in the nucleus. Studies in wheat (Triticum aestivum) have shown that the Pairing homoeologous 1 (Ph1) locus is required to suppress interactions between genetically similar homoeologous chromosomes. Here we show that absence of Ph1 causes increased transcription of Asynapsis 1 (ASY1), a gene that encodes an axial-element-associated protein that is essential for synapsis and cross-over formation in Arabidopsis and rice. Localisation of ASY1 during meiosis is also affected by deletion of Ph1. In addition, transgenic wheat mutants with decreased activity of TaASY1 display reduced synapsis during prophase I and exhibit pairing between homoeologous chromosomes at metaphase I. These results suggest that ASY1 is required to promote interactions between homologous chromosomes in bread wheat, and that Ph1 has a gene regulatory role, which is consistent with its suggested genetic identity as a Cdk-like gene. Broader implications of this research suggest that we could use the Taasy1 mutants to assess their efficacy in alien chromatin introgression studies, as seen with the ph1b mutant.

Journal ArticleDOI
TL;DR: This review explores recent studies addressing the structural and functional characterisation of CenH3‐chromatin, its assembly and propagation during mitosis, and its contribution to kinetochore assembly.
Abstract: The centromere is a specialised chromosomal structure that regulates faithful chromosome segregation during cell division, as it dictates the site of assembly of the kinetochore, a critical structure that mediates binding of chromosomes to the spindle, monitors bipolar attachment and pulls chromosomes to the poles during anaphase. Identified more than a century ago as the primary constriction of condensed metaphase chromosomes, the centromere remained elusive to molecular characterisation for many years owed to its unusual enrichment in highly repetitive satellite DNA sequences, except in budding yeast. In the last decade, our understanding of centromere structure, organisation and function has increased tremendously. Nowadays, we know that centromere identity is determined epigenetically by the formation of a unique type of chromatin, which is characterised by the presence of the centromere-specific histone H3 variant CenH3, originally called CENP-A, which replaces canonical histone H3 at centromeres. CenH3-chromatin constitutes the physical and functional foundation for kinetochore assembly. This review explores recent studies addressing the structural and functional characterisation of CenH3-chromatin, its assembly and propagation during mitosis, and its contribution to kinetochore assembly.

Journal ArticleDOI
TL;DR: This study identifies SKAP as a novel regulator of the metaphase-to-anaphase transition and demonstrates that mis-regulation of the separase activation results in a reduced fidelity of chromosome segregation and a reduced genomic stability independent of the SAC.
Abstract: The spindle assembly checkpoint (SAC) controls the anaphase onset by preventing premature chromosome segregation until bipolar microtubule (MT) attachment and inter-kinetochore tension are fully established for every kinetochore pair. Once the SAC is off, activation of the Anaphase-Promoting Complex/Cyclosome, a ubiquitin ligase, leads to the degradation of securin and cyclin B, the activation of separase and the initiation of anaphase. We report here the identification and characterization of a G(2)-induced gene, SKAP, as a regulator for the anaphase onset. SKAP localizes to spindle MTs and kinetochores in mitosis. Depletion of SKAP does not activate the SAC, but substantially increases the duration of metaphase, delays the activation of separase, and decreases the fidelity of chromosome segregation. Our study identifies SKAP as a novel regulator of the metaphase-to-anaphase transition and demonstrates that misregulation of the separase activation results in a reduced fidelity of chromosome segregation and a reduced genomic stability independent of the SAC.

Journal ArticleDOI
TL;DR: Two novel functions of GAK are reported: maintenance of proper centrosome maturation and of mitotic chromosome congression, which conclude that GAK and clathrin function cooperatively not only in endocytosis, but also in mitotic progression.
Abstract: Cyclin G-associated kinase (GAK) is an association partner of clathrin heavy chain (CHC) and is essential for clathrin-mediated membrane trafficking. Here, we report two novel functions of GAK: maintenance of proper centrosome maturation and of mitotic chromosome congression. Indeed, GAK knockdown by siRNA caused cell-cycle arrest at metaphase, which indicates that GAK is required for proper mitotic progression. We found that this impaired mitotic progression was due to activation of the spindle-assembly checkpoint, which senses protruded, misaligned or abnormally condensed chromosomes in GAK-siRNA-treated cells. GAK knockdown also caused multi-aster formation, which was due to abnormal fragmentation of pericentriolar material, but not of the centrioles. Moreover, GAK and CHC cooperated in the same pathway and interacted in mitosis to regulate the formation of a functional spindle. Taken together, we conclude that GAK and clathrin function cooperatively not only in endocytosis, but also in mitotic progression.

Journal ArticleDOI
TL;DR: Results show that isoliquiritigenin inhibits topoiosmerase II activity, and the resultant DNA damage and arrest in mitotic metaphase-like stage contributes to the antiproliferative effects of isoliquircigenin.