首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Corrigendum     
FBW7 (F-box and WD repeat domain containing 7), also known as FBXW7 or hCDC4, is a tumor suppressor gene mutated in a broad spectrum of cancer cell types. As a component of the SCF E3 ubiquitin ligase, FBW7 is responsible for specifically recognizing phosphorylated substrates, many important for tumor progression, and targeting them for ubiquitin-mediated degradation. Although the role of FBW7 as a tumor suppressor is well established, less well studied is how FBW7-mutated cancer cells might be targeted for selective killing. To explore this further, we undertook a genome-wide RNAi screen using WT and FBW7 knockout colorectal cell lines and identified the spindle assembly checkpoint (SAC) protein BUBR1, as a candidate synthetic lethal target. We show here that asynchronous FBW7 knockout cells have increased levels of mitotic APC/C substrates and are sensitive to knockdown of not just BUBR1 but BUB1 and MPS1, other known SAC components, suggesting a dependence of these cells on the mitotic checkpoint. Consistent with this dependence, knockdown of BUBR1 in cells lacking FBW7 results in significant cell aneuploidy and increases in p53 levels. The FBW7 substrate cyclin E was necessary for the genetic interaction with BUBR1. In contrast, the establishment of this dependence on the SAC requires the deregulation of multiple substrates of FBW7. Our work suggests that FBW7 knockout cells are vulnerable in their dependence on the mitotic checkpoint and that this may be a good potential target to exploit in FBW7-mutated cancer cells.  相似文献   

2.
In this work, we identify physical and genetic interactions that implicate E3 identified by differential display (EDD) in promoting spindle assembly checkpoint (SAC) function. During mitosis, the SAC initiates a mitotic checkpoint in response to chromosomes with kinetochores unattached to spindle pole microtubules. Similar to Budding uninhibited by benzimidazoles-related 1 (BUBR1) siRNA, a bona fide SAC component, EDD siRNA abrogated G2/M accumulation in response to the mitotic destabilizing agent nocodazole. Furthermore, EDD siRNA reduced mitotic cell viability and, in nocodazole-treated cells, increased expression of the promitotic progression protein cell division cycle 20 (CDC20). Copurification studies also identified physical interactions with CDC20, BUBR1, and other components of the SAC. Taken together, these observations highlight the potential role of EDD in regulating mitotic progression and the cellular response to perturbed mitosis.  相似文献   

3.
The multidomain protein kinases BUB1 and BUBR1 (Mad3 in yeast, worms and plants) are central components of the mitotic checkpoint for spindle assembly (SAC). This evolutionarily conserved and essential self-monitoring system of the eukaryotic cell cycle ensures the high fidelity of chromosome segregation by delaying the onset of anaphase until all chromosomes are properly bi-oriented on the mitotic spindle. Despite their amino acid sequence conservation and similar domain organization, BUB1 and BUBR1 perform different functions in the SAC. Recent structural information provides crucial molecular insights into the regulation and recognition of BUB1 and BUBR1, and a solid foundation to dissect the roles of these proteins in the control of chromosome segregation in normal and oncogenic cells.  相似文献   

4.
The Spindle Assembly Checkpoint (SAC) delays the onset of anaphase in response to unattached kinetochores by inhibiting the activity of the Anaphase-Promoting Complex/Cyclosome (APC/C), an E3 ubiquitin ligase. Once all the chromosomes have bioriented, SAC signalling is somehow silenced, which allows progression through mitosis. Recent studies suggest that the APC/C itself participates in SAC silencing by targeting an unknown factor for proteolytic degradation. Key evidence in favour of this model comes from the use of proTAME, a small molecule inhibitor of the APC/C. In cells, proTAME causes a mitotic arrest that is SAC-dependent. Even though this observation comes at odds with the current view that the APC/C acts downstream of the SAC, it was nonetheless argued that these results revealed a role for APC/C activity in SAC silencing. However, we show here that the mitotic arrest induced by proTAME is due to the induction of cohesion fatigue, a phenotype that is caused by the loss of sister chromatid cohesion following a prolonged metaphase. Under these conditions, the SAC is re-activated and APC/C inhibition is maintained independently of proTAME. Therefore, these results provide a simpler explanation for why the proTAME-induced mitotic arrest is also dependent on the SAC. While these observations question the notion that the APC/C is required for SAC silencing, we nevertheless show that APC/C activity does partially contribute to its own release from inhibitory complexes, and importantly, this does not depend on proteasome-mediated degradation.  相似文献   

5.
The mitotic checkpoint maintains genomic stability by ensuring that chromosomes are accurately segregated during mitosis. When the checkpoint is activated, the mitotic checkpoint complex (MCC), assembled from BUBR1, BUB3, CDC20, and MAD2, directly binds and inhibits the anaphase-promoting complex/cyclosome (APC/C) until all chromosomes are properly attached and aligned. The mechanisms underlying MCC assembly and MCC-APC/C interaction are not well characterized. Here, we show that a novel interaction between BUBR1 and closed MAD2 (C-MAD2) is essential for MCC-mediated inhibition of APC/C. Intriguingly, Arg(133) and Gln(134) in C-MAD2 are required for BUBR1 interaction. The same residues are also critical for MAD2 dimerization and MAD2 binding to p31(comet), a mitotic checkpoint silencing protein. Along with previously characterized BUBR1-CDC20 and C-MAD2-CDC20 interactions, our results underscore the integrity of the MCC for its activity and suggest the fundamental importance of the MAD2 αC helix in modulating mitotic checkpoint activation and silencing.  相似文献   

6.
All together now     
Maintenance of genomic stability during eukaryotic cell division relies on the Spindle Assembly Checkpoint (SAC), which has evolved as a surveillance mechanism that monitors kinetochore-microtubule attachment and prevents APC/C-mediated mitotic exit until all chromosomes are properly attached to the mitotic spindle. Reversible protein phosphorylation has long been accredited as a regulatory mechanism of the SAC. Nevertheless, knowledge of how several mitotic kinases act in concert within the signaling pathway to orchestrate SAC function is still emerging. In a recent study, we undertook a comprehensive dissection of the hierarchical framework controlling SAC function in Drosophila cells. We found that Polo lies at the top of the SAC pathway promoting the efficient recruitment of Mps1 to unattached kinetochores. This renders Mps1 fully active to control BubR1 phosphorylation that generates the 3F3/2 phosphoepitope at tensionless kinetochores. We have proposed that Polo is required for SAC function and that the molecular outcome of Mps1-dependent 3F3/2 formation is to promote the association of Cdc20 with BubR1 allowing proper kinetochore recruitment of Cdc20 and efficient assembly of the Mitotic Checkpoint Complex (MCC) required for a sustained SAC response.  相似文献   

7.
MPS1 kinase is an essential component of the spindle assembly checkpoint (SAC), but its functioning mechanisms are not fully understood. We have shown recently that direct interaction between BUBR1 and MAD2 is critical for assembly and function of the human mitotic checkpoint complex (MCC), the SAC effector. Here we report that inhibition of MPS1 kinase activity by reversine disrupts BUBR1-MAD2 as well as CDC20-MAD2 interactions, causing premature activation of the anaphase-promoting complex/cyclosome. The effect of MPS1 inhibition is likely due to reduction of closed MAD2 (C-MAD2), as expressing a MAD2 mutant (MAD2L13A) that is locked in the C conformation rescued the checkpoint defects. In the presence of reversine, exogenous C-MAD2 does not localize to unattached kinetochores but is still incorporated into the MCC. Contrary to a previous report, we found that sustained MPS1 activity is required for maintaining both the MAD1·C-MAD2 complex and open MAD2 (O-MAD2) at unattached kinetochores to facilitate C-MAD2 production. Additionally, mitotic phosphorylation of BUBR1 is also affected by MPS1 inhibition but seems dispensable for MCC assembly. Our results support the notion that MPS1 kinase promotes C-MAD2 production and subsequent MCC assembly to activate the SAC.  相似文献   

8.
Human BUBR1 is a 120 kDa protein that plays a central role in the spindle assembly checkpoint (SAC), the evolutionary conserved and self-regulatory system of higher organisms that monitors and repairs defects in chromosome segregation in mitotic cells. BUBR1 is organised into several domains, with an N-terminal region responsible for its localisation into the kinetochore, the multi-component proteinaceous network that assembles onto chromosomes upon mitotic entry. We have expressed and purified uniformly-15N/13C N-terminal BUBR1 and assigned backbone and side-chain resonances bound to an unlabelled peptide from the protein Blinkin, an element essential for recruitment of BUBR1 to the kinetochore. These assignments provide insights on the Blinkin interaction interface and form the basis of the three-dimensional structure determination of a BUBR1-Blinkin complex.  相似文献   

9.
The spindle assembly checkpoint (SAC) is the major surveillance system that ensures that sister chromatids do not separate until all chromosomes are correctly bioriented during mitosis. Components of the checkpoint include Mad1, Mad2, Mad3 (BubR1), Bub3, and the kinases Bub1, Mph1 (Mps1), and Aurora B. Checkpoint proteins are recruited to kinetochores when individual kinetochores are not bound to spindle microtubules or not under tension. Kinetochore association of Mad2 causes it to undergo a conformational change, which promotes its association to Mad3 and Cdc20 to form the mitotic checkpoint complex (MCC). The MCC inhibits the anaphase-promoting complex/cyclosome (APC/C) until the checkpoint is satisfied. SAC silencing derepresses Cdc20-APC/C activity. This triggers the polyubiquitination of securin and cyclin, which promotes the dissolution of sister chromatid cohesion and mitotic progression. We, and others, recently showed that association of PP1 to the Spc7/Spc105/KNL1 family of kinetochore proteins is necessary to stabilize microtubule-kinetochore attachments and silence the SAC. We now report that phosphorylation of the conserved MELT motifs in Spc7 by Mph1 (Mps1) recruits Bub1 and Bub3 to the kinetochore and that this is required to maintain the SAC signal.  相似文献   

10.
The mitotic checkpoint is a specialized signal transduction pathway that monitors kinetochore-microtubule attachment to achieve faithful chromosome segregation. MAD2 is an evolutionarily conserved mitotic checkpoint protein that exists in open (O) and closed (C) conformations. The increase of intracellular C-MAD2 level during mitosis, through O?C-MAD2 conversion as catalyzed by unattached kinetochores, is a critical signaling event for the mitotic checkpoint. However, it remains controversial whether MAD2 is an integral component of the effector of the mitotic checkpoint---the Mitotic Checkpoint Complex (MCC). We show here that endogenous human MCC is assembled by first forming a BUBR1:BUB3:CDC20 complex in G2 and then selectively incorporating C-MAD2 during mitosis. Nevertheless, MCC can be induced to form in G1/S cells by expressing a C-conformation locked MAD2 mutant, indicating intracellular level of C-MAD2 as a major limiting factor for MCC assembly. In addition, a recombinant MCC containing C-MAD2 exhibits effective inhibitory activity towards APC/C isolated from mitotic HeLa cells, while a recombinant BUBR1:BUB3:CDC20 ternary complex is ineffective at comparable concentrations despite association with APC/C. These results help establish a direct connection between a major signal transducer (C-MAD2) and the potent effector (MCC) of the mitotic checkpoint, and provide novel insights into protein-protein interactions during assembly of a functional MCC.  相似文献   

11.
The E6 and E7 genes of the high-risk human papillomavirus (HPV) types encode oncoproteins, and both act by interfering with the activity of cellular tumor suppressor proteins. E7 proteins act by associating with members of the retinoblastoma family, while E6 increases the turnover of p53. p53 has been implicated as a regulator of both the G1/S cell cycle checkpoint and the mitotic spindle checkpoint. When fibroblasts from p53 knockout mice are treated with the spindle inhibitor nocodazole, a rereplication of DNA occurs without transit through mitosis. We investigated whether E6 or E7 could induce a similar loss of mitotic checkpoint activity in human keratinocytes. Recombinant retroviruses expressing high-risk E6 alone, E7 alone, and E6 in combination with E7 were used to infect normal human foreskin keratinocytes (HFKs). Established cell lines were treated with nocodazole, stained with propidium iodide, and analyzed for DNA content by flow cytometry. Cells infected with high-risk E6 were found to continue to replicate DNA and accumulated an octaploid (8N) population. Surprisingly, expression of E7 alone was also able to bypass this checkpoint. Cells expressing E7 alone exhibited increased levels of p53, while those expressing E6 had significantly reduced levels. The p53 present in the E7 cells was active, as increased levels of p21 were observed. This suggested that E7 bypassed the mitotic checkpoint by a p53-independent mechanism. The levels of MDM2, a cellular oncoprotein also implicated in control of the mitotic checkpoint, were significantly elevated in the E7 cells compared to the normal HFKs. In E6-expressing cells, the levels of MDM2 were undetectable. It is possible that abrogation of Rb function by E7 or increased expression of MDM2 contributes to the loss of mitotic spindle checkpoint control in the E7 cells. These findings suggest mechanisms by which both HPV oncoproteins contribute to genomic instability at the mitotic checkpoint.  相似文献   

12.
在有丝分裂过程中BUBR1监视微管与着丝点的结合,是保证染色体均等分离的重要分子机制之一.BUBIB变异家谱研究及其敲除模型的研究表明,BUBR1缺陷与染色体不稳定性及肿瘤的发生直接相关.近来在数种人类肿瘤,对BUBR1蛋白过度表达有所报道.但在直结肠癌,BUBR1的过度表达是否与染色体不稳定性的发生有关目前仍无定论.在人类结直肠癌的遗传不稳定性主要表现为两种类型,染色体不稳定性及微卫星不稳定性,它们提示了两条独立的肿瘤发生路径.一般认为不存在高频度微卫星不稳定性表型的肿瘤通过染色体不稳定途径癌变.P53蛋白通过多种机制对维护遗传稳定性起到重要的作用,TP53基因突变经常与染色体不稳定现象并存.DNA倍体情况也是染色体不稳定研究不可缺少的指标.本研究采用免疫组织化学法检测了一组93例进展期散发结直肠癌BUBR1蛋白的表达情况,直接测序法检测TP53变异.高分辨率荧光标记微卫星不稳定检测技术检测微卫星状态,固相激光扫描细胞仪技术检测DNA倍体情况.我们分析了BUBR1表达与三种反映遗传背景的因子的关系.BUBR1蛋白过度表达在人结直肠癌较为常见.在非高频度微卫星不稳定的结直肠癌,BUBR1蛋白过度表达率明显为高(P<0.01),在TP53基因突变的病例其过度表达率亦较高(P<0.05).BUBR1蛋白的过度表达与DNA异倍体无统计学相关,但DNA异倍体病例的BuBRl过度表达有偏高倾向.BuBRl表达情况与常用的临床病理学指标无统计学相关.BuBRl过度表达同微卫星状态及TP53突变的关系明确的提示,在人类散发结直肠癌,BUBR1蛋白过度表达与染色体不稳定状态有关.BUBR1过度表达作为一种常见的分子异常,对于肿瘤的早诊预防提供新的标志物.并可能成为治疗的新靶点.  相似文献   

13.
Silencing of the spindle assembly checkpoint involves two protein phosphatases, PP1 and PP2A-B56, that are thought to extinguish checkpoint signaling through dephosphorylation of a checkpoint scaffold at kinetochores. In this issue, Cordeiro et al. (2020. J. Cell Biol. https://doi.org/10.1083/jcb.202002020) now show that a critical function of these phosphatases in checkpoint silencing is removal of Polo kinase at kinetochores, which would otherwise autonomously sustain the checkpoint.

The main goal of mitosis is to accurately segregate chromosomes, such that each daughter cell inherits a full complement of genetic information. To accomplish this delicate task, once each chromosome is faithfully duplicated through DNA replication, its identical sister chromatids must attach to spindle microtubules coming from opposite spindle poles through a process known as chromosome biorientation. Kinetochores are proteinaceous assemblies that reside at the centromeric region of chromosomes and are key to this process by capturing spindle microtubules (1). Microtubule capture, however, is inherently error prone, and several cycles of attachment/detachment are often required before chromosomes achieve biorientation. Obviously, chromosome segregation without error correction would be highly detrimental, leading to unbalanced chromosome numbers, referred to as aneuploidies, which are hallmarks of cancer and genetic diseases. Luckily, eukaryotic cells not only possess an error-correction machinery deputed to rectify faulty attachments (2), but they also have a safeguard device, called the spindle assembly checkpoint (SAC), that temporarily halts cells in mitosis to provide them with the necessary time window to fix the errors. SAC signaling fires at unattached kinetochores, which are continuously generated during error correction, and is extinguished once all chromosomes are bioriented, thus resuming mitotic progression and chromosome segregation (3).Prevailing models posit that a key trigger of SAC signaling is the phosphorylation of the kinetochore scaffold KNL1 by the SAC kinase MPS1. This creates a phospho-docking site at the MELT repeats (amino acid consensus Met-Glu-Leu-Thr) of KNL1 that recruits the heterotetrameric SAC complex BUB1:BUB3:BUB3:BUBR1 (referred to as BUB complex; Fig. 1), which in turn attracts to the kinetochore other SAC proteins that collectively prevent mitotic progression (3).Open in a separate windowFigure 1.The interplay of SAC kinases and phosphatases at kinetochores. When SAC is activated at an unattached kinetochore (SAC on), MPS1 phosphorylates the kinetochore scaffold KNL1, thereby recruiting the BUB complex. Contextually, CDK1-dependent phosphorylation of BUB1 and BUBR1 generates phospho-docking sites for recruitment of the Polo kinase PLK1, which on one side sustains KNL1 phosphorylation and on the other stimulates BUBR1 binding to PP2A-B56. The latter, in turn, counteracts PLK1 local activity by dislodging PLK1 from the kinetochore. During SAC silencing, local activity of MPS1 is shut off. Additionally, the PP1 phosphatase binds to KNL1 and, together with PP2A-B56, further evicts PLK1 from the kinetochore, possibly through dephosphorylation of its phospho-docking sites in BUB1 and BUBR1. This leads to KNL1 dephosphorylation and displacement of the BUB complex, thus extinguishing SAC signaling (SAC off). Whether PP1 and PP2A-B56, as opposed to other phosphatases, contribute directly to KNL1 dephosphorylation remains an open question.The protein phosphatases PP1 and PP2A-B56 are recruited to kinetochores through binding to KNL1 and BUBR1, respectively, and are thought to silence the SAC through dephosphorylation of the MELT repeats of KNL1, thus antagonizing MPS1 activity (Fig. 1). Additional mechanisms, such as MPS1 inhibition and stripping of SAC components from kinetochores, have been proposed to contribute to obliterate SAC signaling upon chromosome biorientation (4).In this issue, compelling evidence from Cordeiro et al. challenges the current view by showing that rather than dephosphorylating KNL1, PP1 and PP2A-B56 actually silence the SAC by down-regulating the activity of Polo kinase (PLK1 in human cells) at kinetochores (5). Polo kinase and MPS1 share a common substrate preference and both can phosphorylate the MELT repeats of KNL1. Additionally, Polo cooperates with MPS1 in SAC signaling in various species, while in organisms where MPS1 is absent, like nematodes, Polo functionally replaces MPS1 (6).Consistent with previous results (7, 8), Cordeiro et al. show that when kinetochore phosphatases are dampened, PLK1 levels increase at kinetochores through an unknown mechanism, which might involve dephosphorylation of the phosphoepitopes in the Polo-binding motifs generated on the BUB complex by CDK1 (BUB1-pT609 and BUBR1-pT620; 9, 10, 11). This implies that when PP1 and PP2A-B56 are low at kinetochores, PLK1 can amplify SAC signaling through a positive feedback loop by boosting KNL1 phosphorylation independently of Mps1, thereby recruiting the BUB complex and, in turn, increasing amounts of PLK1 (Fig. 1). In agreement with this view, in a sensitized setup where kinetochore phosphatases are crippled along with MPS1, concomitant inhibition of PLK1 is sufficient to bring about KNL1 dephosphorylation and restore SAC signaling. These data led the authors to the provocative conclusion that the primary role of PP1 and PP2A-B56 in SAC silencing is to harness PLK1 activity. This new model is appealing not only because it highlights a novel function for PP1 and PP2A-B56 in SAC silencing, but also because it explains the modest effects that are commonly observed on SAC signaling upon PLK1 inhibition alone. Indeed, kinetochore phosphatases, and primarily PP2A-B56 (12), are already partially active in a SAC-induced mitotic arrest (e.g., upon microtubule depolymerization), as shown here by the increased KNL1 phosphorylation upon their inactivation.Interestingly, sequence alignment of BUB1 and BUBR1 homologues across the phylogenetic tree reveals that, in metazoans, putative Polo-binding motifs are usually located in the vicinity of hypothetical PP2A-B56–binding motifs, suggesting that they coevolved. The physical proximity of Polo-binding and PP2A-B56–binding motifs in BUB1 and BUBR1 could position the Polo-binding motifs in a favorable arrangement for their PP2A-B56–driven dephosphorylation and, as a consequence, PLK1 clearance from kinetochores (Fig. 1).The data by Cordeiro et al. represent a paradigm shift in our understanding of SAC silencing for two main reasons. First, consistent with published data (13, 14), PP1 and PP2A-B56 might be involved in this process primarily by inactivating upstream SAC kinases (MPS1 and PLK1), rather than dephosphorylating their substrates. Second, since PLK1 is partially displaced from kinetochores by the above phosphatases already during a SAC arrest, MPS1 inactivation might be the main trigger of SAC silencing. Several mechanisms have been proposed to attenuate MPS1 activity once the SAC is satisfied, such as MPS1 displacement from kinetochores (6) and dephosphorylation of MPS1 in its activation loop (13, 14).The new model raises a burning question: If PP1 and PP2A do not dephosphorylate KNL1 at MELT repeats, what does? Other phosphatases, whose identity remains elusive, could be involved in KNL1 dephosphorylation. Alternatively, phosphorylated KNL1 might be actively turned over at kinetochores. Nevertheless, at present, the involvement of PP1 and PP2A-B56 in KNL1 dephosphorylation cannot be ruled out, as complete inhibition of kinetochore phosphatases in the experimental setup used here is likely very challenging. Further investigations will be required to solve this central issue.Another important question that deserves further scrutiny is, how exactly do PP1 and PP2A-B56 inhibit PLK1 activity at kinetochores? Cordeiro et al. propose that they could dephosphorylate the Polo-binding motifs in BUB1/BUBR1. Alternatively, the close proximity of PP2A- and Polo-binding motifs in metazoan BUBR1 homologues could make the association of PLK1 and PP2A with BUBR1 mutually exclusive.Finally, and most importantly, what is the physiological meaning of the complex interplay between SAC kinases and phosphatases described here? A crucial function of PLK1 bound to the BUB complex in human cells is to stabilize kinetochore-microtubule attachments in prometaphase by recruiting PP2A-B56 through phosphorylation of the PP2A-B56-binding motif in BUBR1 (15). In turn, eviction of PLK1 from kinetochores by PP2A-B56 will have two major outputs: (i) maintain microtubule dynamics at bioriented chromosomes (8) and (ii) stimulate binding of PP1 to KNL1, which primes the system for SAC silencing (16). As soon as MPS1 levels drop at kinetochores and/or other phosphatases intervene to dephosphorylate KNL1, SAC signaling is finally extinguished. The development of fluorescence-based biosensors combined with mathematical modeling will certainly provide in the future further mechanistic insights into such intricate network.  相似文献   

14.
Cancer cells contain an abnormal number of chromosomes (aneuploidy), which is a prevalent form of genetic instability in human cancers. Abnormal amplification of centrosomes and defects of spindle assembly checkpoint are the major causes of chromosome instability in cancer cells. Here we present biochemical evidence to suggest a role of ECRG2, a novel tumor suppressor gene, in maintaining chromosome stability. ECRG2 localized to centrosomes during interphase and kinetochores during mitosis. Further analysis revealed that ECRG2 participates in centrosome amplification in a p53-dependent manner. Depletion of ECRG2 not only destabilized p53, down-regulated p21, and increased the cyclin E/CDK2 activity, thus initiating centrosome amplification, but also abolished the ability of p53 localize to centrosomes. Overexpression of ECRG2 restored the p53-dependent suppression of centrosome duplication. Furthermore, ECRG2-depleted cells show severely disrupted spindle phenotype but fail to maintain the mitotic arrest due to minimal BUBR1 protein levels. Taken together, our results indicate that ECRG2 is important for ensuring centrosome duplication, spindle assembly checkpoint, and accurate chromosome segregation, and its depletion may contribute to chromosome instability and aneuploidy in human cancers.  相似文献   

15.
16.
Eukaryotic cells have evolved a mechanism that delays the progression of mitosis until condensed chromosomes are properly positioned on the mitotic spindle. To understand the molecular basis of such monitoring mechanism in human cells, we have been studying genes that regulate the mitotic checkpoint. Our early studies have led to the cloning of a full-length cDNA encoding MAD3-like protein (also termed BUBR1/MAD3/SSK1). Dot blot analyses show that BUBR1 mRNA is expressed in tissues with a high mitotic index but not in differentiated tissues. Western blot analyses show that in asynchronous cells, BUBR1 protein primarily exhibits a molecular mass of 120 kDa, and its expression is detected in most cell lines examined. In addition, BUBR1 is present during various stages of the cell cycle. As cells enter later S and G2, BUBR1 levels are increased significantly. Nocodazole-arrested mitotic cells obtained by mechanical shake-off contain BUBR1 antigen with a slower mobility on denaturing SDS gels. Phosphatase treatment restores the slowly migrating band to the interphase state, indicating that the slow mobility of the BUBR1 antigen is attributable to phosphorylation. Furthermore, purified recombinant His6-BUBR1 is capable of autophosphorylation. Our studies indicate that BUBR1 phosphorylation status is regulated during spindle disruption. Considering its strong homology to BUB1 protein kinase, BUBR1 may also play an important role in mitotic checkpoint control by phosphorylation of a critical cellular component(s) of the mitotic checkpoint pathway.  相似文献   

17.
The maintenance of genomic stability relies on the spindle assembly checkpoint (SAC), which ensures accurate chromosome segregation by delaying the onset of anaphase until all chromosomes are properly bioriented and attached to the mitotic spindle. BUB1 and BUBR1 kinases are central for this process and by interacting with Blinkin, link the SAC with the kinetochore, the macromolecular assembly that connects microtubules with centromeric DNA. Here, we identify the Blinkin motif critical for interaction with BUBR1, define the stoichiometry and affinity of the interaction, and present a 2.2 ? resolution crystal structure of the complex. The structure defines an unanticipated BUBR1 region responsible for the interaction and reveals a novel Blinkin motif that undergoes a disorder-to-order transition upon ligand binding. We also show that substitution of several BUBR1 residues engaged in binding Blinkin leads to defects in the SAC, thus providing the first molecular details of the recognition mechanism underlying kinetochore-SAC signaling.  相似文献   

18.
The spindle assembly checkpoint (SAC) restrains anaphase until all chromosomes become bi-oriented on the mitotic spindle. The SAC protein Mad2 can fold into two distinct conformers, open (O) and closed (C), and can asymmetrically dimerize. Here, we describe a monoclonal antibody that specifically recognizes the dimerization interface of C-Mad2. This antibody revealed several conformation-specific features of Mad2 in human cells. Notably, we show that Mad2 requires association with Mad1 to adopt the closed conformation and that the activity of the Mad1:C-Mad2 complex undergoes regulation by p31comet-dependent 'capping'. Furthermore, C-Mad2 antibody microinjection caused an abrupt termination of the SAC and accelerated mitotic progression. Remarkably, microinjection of a Mad1-neutralizing antibody triggered a comparable mitotic acceleration. Our study provides direct in vivo evidence for the model that a kinetochore complex of Mad1:C-Mad2 acts as a template to sustain the SAC and it challenges the distinction between SAC and mitotic timer.  相似文献   

19.
The centromere/kinetochore complex plays an essential role in cell and organismal viability by ensuring chromosome movements during mitosis and meiosis. The kinetochore also mediates the spindle attachment checkpoint (SAC), which delays anaphase initiation until all chromosomes have achieved bipolar attachment of kinetochores to the mitotic spindle. CENP-A proteins are centromere-specific chromatin components that provide both a structural and a functional foundation for kinetochore formation. Here we show that cells in Drosophila embryos homozygous for null mutations in CENP-A (CID) display an early mitotic delay. This mitotic delay is not suppressed by inactivation of the DNA damage checkpoint and is unlikely to be the result of DNA damage. Surprisingly, mutation of the SAC component BUBR1 partially suppresses this mitotic delay. Furthermore, cid mutants retain an intact SAC response to spindle disruption despite the inability of many kinetochore proteins, including SAC components, to target to kinetochores. We propose that SAC components are able to monitor spindle assembly and inhibit cell cycle progression in the absence of sustained kinetochore localization.  相似文献   

20.
Aneuploidy and chromosomal instability (CIN) are common features of gastric cancer (GC), but their contribution to carcinogenesis and antitumour therapy response is still poorly understood. Failures in the mitotic checkpoint induced by changes in expression levels of the spindle assembly checkpoint (SAC) proteins cause the missegregation of chromosomes in mitosis as well as aneuploidy. To evaluate the possible contribution of SAC to GC, we analyzed the expression levels of proteins of the mitotic checkpoint complex in a cohort of GC cell lines. We found that the central SAC proteins, Mad2 and BubR1, were the more prominently expressed members in disseminated GC cell lines. Silencing of Mad2 and BubR1 in MKN45 and ST2957 cells decreased their cell proliferation, migration and invasion abilities, indicating that Mad2 and BubR1 could contribute to cellular transformation and tumor progression in GC. We next evaluated whether silencing of SAC proteins could affect the response to microtubule poisons. We discovered that paclitaxel treatment increased cell survival in MKN45 cells interfered for Mad2 or BubR1 expression. However, apoptosis (assessed by caspase-3 activation, PARP proteolysis and levels of antiapoptotic Bcl 2-family members), the DNA damage response (assessed by H2Ax phosphorylation) and exit from mitosis (assessed by Cyclin B degradation and Cdk1 regulation) were activated equally between cells, independently of Mad2 or BubR1-protein levels. In contrast, we observed that the silencing of Mad2 or BubR1 in MKN45 cells showed the induction of a senescence-like phenotype accompanied by cell enlargement, increased senescence-associated β-galactosidase activity and increased IL-6 and IL-8 expression. In addition, the senescent phenotype is highly increased after treatment with PTX, indicating that senescence could prevent tumorigenesis in GC. In conclusion, the results presented here suggest that Mad2 and BubR1 could be used as prognostic markers of tumor progression and new pharmacological targets in the treatment for GC.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号