340 results on '"BUBR1"'
Search Results
2. BUB1B monoallelic germline variants contribute to prostate cancer predisposition by triggering chromosomal instability.
- Author
-
Silva, Maria P., Ferreira, Luísa T., Brás, Natércia F., Torres, Lurdes, Brandão, Andreia, Pinheiro, Manuela, Cardoso, Marta, Resende, Adriana, Vieira, Joana, Palmeira, Carlos, Martins, Gabriela, Silva, Miguel, Pinto, Carla, Peixoto, Ana, Silva, João, Henrique, Rui, Maia, Sofia, Maiato, Helder, Teixeira, Manuel R., and Paulo, Paula
- Subjects
- *
HEREDITARY cancer syndromes , *PROSTATE cancer , *GENETIC testing , *SPINDLE apparatus , *GERM cells , *GENETIC counseling - Abstract
Background: Prostate cancer (PrCa) is the most frequently diagnosed cancer in men. Variants in known moderate- to high-penetrance genes explain less than 5% of the cases arising at early-onset (< 56 years) and/or with familial aggregation of the disease. Considering that BubR1 is an essential component of the mitotic spindle assembly checkpoint, we hypothesized that monoallelic BUB1B variants could be sufficient to fuel chromosomal instability (CIN), potentially triggering (prostate) carcinogenesis. Methods: To unveil BUB1B as a new PrCa predisposing gene, we performed targeted next-generation sequencing in germline DNA from 462 early-onset/familial PrCa patients and 1,416 cancer patients fulfilling criteria for genetic testing for other hereditary cancer syndromes. To explore the pan-cancer role of BUB1B, we used in silico BubR1 molecular modeling, in vitro gene-editing, and ex vivo patients' tumors and peripheral blood lymphocytes. Results: Rare BUB1B variants were found in ~ 1.9% of the early-onset/familial PrCa cases and in ~ 0.6% of other cancer patients fulfilling criteria for hereditary disease. We further show that BUB1B variants lead to decreased BubR1 expression and/or stability, which promotes increased premature chromatid separation and, consequently, triggers CIN, driving resistance to Taxol-based therapies. Conclusions: Our study shows that different BUB1B variants may uncover a trigger for CIN-driven carcinogenesis, supporting the role of BUB1B as a (pan)-cancer predisposing gene with potential impact on genetic counseling and treatment decision-making. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Overexpression of BubR1 Mitotic Checkpoint Protein Predicts Short Survival and Influences the Progression of Cholangiocarcinoma.
- Author
-
Pokaew, Nongnapas, Prajumwongs, Piya, Vaeteewoottacharn, Kulthida, Wongkham, Sopit, Pairojkul, Chawalit, and Sawanyawisuth, Kanlayanee
- Subjects
OVERALL survival ,CELL lines ,CHOLANGIOCARCINOMA ,CELL proliferation ,SURVIVAL rate - Abstract
Budding Uninhibited by Benzimidazole-Related 1 (BubR1) or BUB1 Mitotic Checkpoint Serine/Threonine Kinase B (BUB1B) is an essential component of the spindle assembly checkpoint (SAC), which controls chromosome separation during mitosis. Overexpression of BubR1 has been associated with the progression of various cancers. This study demonstrated that high expression of BubR1 correlated with cholangiocarcinogenesis in a hamster cholangiocarcinoma (CCA) model and was associated with shorter survival in patients with CCA. Co-expression of BubR1 and MPS1, which is a SAC-related protein, indicated a shorter survival rate in patients with CCA. Knockdown of BubR1 expression by specific siRNA (siBubR1) significantly decreased cell proliferation and colony formation while inducing apoptosis in CCA cell lines. In addition, suppression of BubR1 inhibited migration and invasion abilities via epithelial–mesenchymal transition (EMT). A combination of siBubR1 and chemotherapeutic drugs showed synergistic effects in CCA cell lines. Taken together, this finding suggested that BubR1 had oncogenic functions, which influenced CCA progression. Suppression of BubR1 might be an alternative option for CCA treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. SETDB2 interacts with BUBR1 to induce accurate chromosome segregation independently of its histone methyltransferase activity
- Author
-
Yanhong Tu, Haomiao Zhang, Jialin Xia, Yu Zhao, Ruifang Yang, Jing Feng, Xueyun Ma, and Jing Li
- Subjects
APC/C ,BUBR1 ,CDC20 ,chromosome segregation ,mitosis ,SETDB2 ,Biology (General) ,QH301-705.5 - Abstract
SETDB2 is a H3K9 histone methyltransferase required for accurate chromosome segregation. Its H3K9 histone methyltransferase activity was reported to be associated with chromosomes during metaphase. Here, we confirm that SETDB2 is required for mitosis and accurate chromosome segregation. However, these functions are independent of its histone methyltransferase activity. Further analysis showed that SETDB2 can interact with BUBR1, and is required for CDC20 binding to BUBR1 and APC/C complex and CYCLIN B1 degradation. The ability of SETDB2 to regulate the binding of CDC20 to BUBR1 or APC/C complex, and stabilization of CYCLIN B1 are also independent of its histone methyltransferase activity. These results suggest that SETDB2 interacts with BUBR1 to promote binding of CDC20 to BUBR1 and APC3, then degrades CYCLIN B1 to ensure accurate chromosome segregation and mitosis, independently of its histone methyltransferase activity.
- Published
- 2024
- Full Text
- View/download PDF
5. SETDB2 interacts with BUBR1 to induce accurate chromosome segregation independently of its histone methyltransferase activity.
- Author
-
Tu, Yanhong, Zhang, Haomiao, Xia, Jialin, Zhao, Yu, Yang, Ruifang, Feng, Jing, Ma, Xueyun, and Li, Jing
- Subjects
CHROMOSOME segregation ,METHYLTRANSFERASES ,CENTROMERE ,CELL cycle proteins ,MITOSIS ,CYCLINS - Abstract
SETDB2 is a H3K9 histone methyltransferase required for accurate chromosome segregation. Its H3K9 histone methyltransferase activity was reported to be associated with chromosomes during metaphase. Here, we confirm that SETDB2 is required for mitosis and accurate chromosome segregation. However, these functions are independent of its histone methyltransferase activity. Further analysis showed that SETDB2 can interact with BUBR1, and is required for CDC20 binding to BUBR1 and APC/C complex and CYCLIN B1 degradation. The ability of SETDB2 to regulate the binding of CDC20 to BUBR1 or APC/C complex, and stabilization of CYCLIN B1 are also independent of its histone methyltransferase activity. These results suggest that SETDB2 interacts with BUBR1 to promote binding of CDC20 to BUBR1 and APC3, then degrades CYCLIN B1 to ensure accurate chromosome segregation and mitosis, independently of its histone methyltransferase activity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. E3-ubiquitin ligase, FBXW7 regulates mitotic progression by targeting BubR1 for ubiquitin-mediated degradation.
- Author
-
Nair, Vishnu M., Sabu, Amit Santhu, Hussain, Ahmed, Kombarakkaran, Delvin P., Lakshmi, R. Bhagya, and Manna, Tapas K.
- Abstract
Faithful chromosome segregation requires correct attachment of kinetochores with the spindle microtubules. Erroneously-attached kinetochores recruit proteins to activate Spindle assembly checkpoint (SAC), which senses the errors and signals cells to delay anaphase progression for error correction. Temporal control of the levels of SAC activating-proteins is critical for checkpoint activation and silencing, but its mechanism is not fully understood. Here, we show that E3 ubiquitin ligase, SCF-FBXW7 targets BubR1 for ubiquitin-mediated degradation and thereby controls SAC in human cells. Depletion of FBXW7 results in prolonged metaphase arrest with increased stabilization of BubR1 at kinetochores. Similar kinetochore stabilization is also observed for BubR1-interacting protein, CENP-E. FBXW7 induced ubiquitination of both BubR1 and the BubR1-interacting kinetochore-targeting domain of CENP-E, but CENP-E domain degradation is dependent on BubR1. Interestingly, Cdk1 inhibition disrupts FBXW7-mediated BubR1 targeting and further, phospho-resistant mutation of Cdk1-targeted phosphorylation site, Thr 620 impairs BubR1-FBXW7 interaction and FBXW7-mediated BubR1 ubiquitination, supporting its role as a phosphodegron for FBXW7. The results demonstrate SCF-FBXW7 as a key regulator of spindle assembly checkpoint that controls stability of BubR1 and its associated CENP-E at kinetochores. They also support that upstream Cdk1 specific BubR1 phosphorylation signals the ligase to activate the process. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Downregulation of BUBR1 regulates the proliferation and cell cycle of breast cancer cells and increases the sensitivity of cells to cisplatin.
- Author
-
Lu, Yiran, Wang, Ruiqing, He, Song, Zhang, Qing, Wei, Jiahui, Hu, Jinping, and Ding, Yu
- Abstract
Breast cancer (BC) is a significant tissue for women's health worldwide. The spindle assembly checkpoint protein family includes BUBR1 (Bub1-related kinase or MAD3/Bub1b). High expression of BUBR1 promotes cell cycle disorders, leading to cell carcinogenesis and cancer progression. However, the underlying molecular mechanism and the role of BUBR1 in BC progression are unclear. The published dataset was analyzed to evaluate the clinical relevance of BUBR1. BUBR1 was knocked down in BC cells using shRNA. The CCK-8 assay was used to measure the cell viability, and mRNA and protein expression levels were detected by RT-qPCR and Western blot (WB). Cell apoptosis and cell cycle were detected by flow cytometry. Subcutaneous xenograft model was used to assess in vivo tumor growth. BUBR1 was found to be highly expressed in BC. The high expression of BUBR1 was associated with poor prognosis of BC patients. Upon BUBR1 knockdown using shRNA, the proliferation and metastatic ability of cells were decreased. Moreover, the cells with BUBR1 knockdown underwent cell cycle arrest. And the results showed that BUBR1 loss inhibited the phosphorylation of TAK1/JNK. In vitro and in vivo studies indicated the knockdown of BUBR1 rendered the BC cells more sensitive to cisplatin. In summary, BUBR1 may be a potential therapeutic target for BC and targeting BUBR1 may help overcome cisplatin resistance in BC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Overexpression of BubR1 Mitotic Checkpoint Protein Predicts Short Survival and Influences the Progression of Cholangiocarcinoma
- Author
-
Nongnapas Pokaew, Piya Prajumwongs, Kulthida Vaeteewoottacharn, Sopit Wongkham, Chawalit Pairojkul, and Kanlayanee Sawanyawisuth
- Subjects
BubR1 ,BUB1B ,mitotic checkpoint ,cholangiocarcinoma ,Biology (General) ,QH301-705.5 - Abstract
Budding Uninhibited by Benzimidazole-Related 1 (BubR1) or BUB1 Mitotic Checkpoint Serine/Threonine Kinase B (BUB1B) is an essential component of the spindle assembly checkpoint (SAC), which controls chromosome separation during mitosis. Overexpression of BubR1 has been associated with the progression of various cancers. This study demonstrated that high expression of BubR1 correlated with cholangiocarcinogenesis in a hamster cholangiocarcinoma (CCA) model and was associated with shorter survival in patients with CCA. Co-expression of BubR1 and MPS1, which is a SAC-related protein, indicated a shorter survival rate in patients with CCA. Knockdown of BubR1 expression by specific siRNA (siBubR1) significantly decreased cell proliferation and colony formation while inducing apoptosis in CCA cell lines. In addition, suppression of BubR1 inhibited migration and invasion abilities via epithelial–mesenchymal transition (EMT). A combination of siBubR1 and chemotherapeutic drugs showed synergistic effects in CCA cell lines. Taken together, this finding suggested that BubR1 had oncogenic functions, which influenced CCA progression. Suppression of BubR1 might be an alternative option for CCA treatment.
- Published
- 2024
- Full Text
- View/download PDF
9. Caspase cleaves DrosophilaBubR1 to modulate spindle assembly checkpoint function and lifespan of the organism.
- Author
-
Shinoda, Natsuki, Horikoshi, Misuzu, Taira, Yusuke, Muramoto, Masaya, Hirayama, Shoshiro, Murata, Shigeo, and Miura, Masayuki
- Subjects
- *
PROTEIN-protein interactions , *KINETOCHORE , *DROSOPHILA - Abstract
Caspases cleave over 1500 substrates in the human proteome in both lethal and non‐lethal scenarios. However, reports of the physiological consequences of substrate cleavage are limited. Additionally, the manner in which caspase cleaves only a subset of substrates in the non‐lethal scenario remains to be elucidated. BubR1, a spindle assembly checkpoint component, is a caspase substrate in humans, the physiological function of which remains unclear. Here, we found that caspases, especially Drice, cleave Drosophila BubR1 between the N‐terminal KEN box motif and C‐terminal kinase domain. By using proximity labelling, we found that Drice, but not Dcp‐1, is in proximity to BubR1, suggesting that protein proximity facilitates substrate preference. The cleaved fragments displayed altered subcellular localization and protein–protein interactions. Flies that harboured cleavage‐resistant BubR1 showed longer duration of BubR1 localization to the kinetochore upon colchicine treatment. Furthermore, these flies showed extended lifespan. Thus, we propose that the caspase‐mediated cleavage of BubR1 limits spindle assembly checkpoint and organismal lifespan. Our results highlight the importance of the individual analysis of substrates in vivo to determine the biological significance of caspase‐dependent non‐lethal cellular processes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Molecular insights into kinetochore-microtubule attachments in mitosis
- Author
-
Legal, Thibault, Welburn, Julie, and Hardwick, Kevin
- Subjects
mitosis ,microtubules ,Dam1 ,kinetochore-microtubule attachments ,CENP-E ,BubR1 ,spindle assembly checkpoint ,kinetochore-recruitment pathways - Abstract
During mitosis, cells must ensure that chromosomes are correctly segregated to both daughter cells. Chromosomes must attach microtubules from opposite spindle poles and congress to the metaphase plate. Once all the chromosomes are bioriented, the cell proceeds to anaphase and chromosomes migrate to opposite poles. Microtubules are dynamic filaments that grow and shrink. It is essential that chromosomes maintain their attachment to microtubules during mitosis to allow for correct chromosome segregation. Chromosomes are attached to microtubules of the mitotic spindle via their kinetochore, a multiprotein complex scaffold that assembles onto centromeric DNA. First, I studied the molecular basis of outer kinetochore-microtubule attachments in the budding yeast Saccharomyces cerevisiae. There, the Dam1 complex is an outer kinetochore-localised heterodecamer that maintains correct kinetochore attachment to a single incoming microtubule in vivo. The organisation of the ten different subunits within a single complex and how multiple complexes oligomerise is unclear. Furthermore, which domains of which subunits are responsible for microtubule binding remains under debate. Using cross-linking coupled with mass spectrometry, I shed light on the subunit organisation of Dam1 heterodecamers and obtained information on domains mediating oligomerisation. I demonstrated that the flexible C termini of both Duo1 and Dam1 subunits were essential for microtubule binding by designing mutants and assessing their microtubule-binding abilities. This work shows Duo1 and Dam1 subunits are placed in close proximity to the microtubule lattice and furthers our understanding of the overall structure of the Dam1 complex. I then focussed on kinetochore-microtubule attachments in humans. In higher eukaryotes, chromosomes align along the metaphase plate and biorient via different processes involving motors and microtubules. Some are immediately captured by microtubules while others move to the spindle poles before sliding from the poles to the metaphase plate, following the lattice of a kinetochore fibre. CENP-E is a large kinesin motor essential for chromosome congression. However, how CENP-E is recruited to unattached kinetochores remains poorly defined. Using biochemistry and cell biology, I showed that a minimal kinetochore-targeting domain of CENP-E interacts with the spindle checkpoint protein BubR1 directly in vitro and that the C-terminal helix of the pseudokinase domain of BubR1 is essential but not sufficient for CENP-E binding. Conversely, CENP-E necessitates a conserved acidic patch close to its C terminus for BubR1 binding. This precise mapping of the interaction allowed us to interrogate the role of BubR1 in recruiting CENP-E to kinetochores. We revealed that this interaction is essential for CENP-E recruitment in early mitosis and in metaphase. These experiments also highlight that CENP-E is recruited to kinetochores via another, uncharacterised, pathway. This work led to the third aim of my thesis: identify an alternative recruitment pathway of CENP-E to kinetochores. CENP-E localises to the outer corona of kinetochores. Interestingly, CENP-E detaches from kinetochores after a short treatment with a CDK1 inhibitor. CENP-E is then found in a protein complex that contains other detached corona proteins such as the RZZ complex, the Dynein adaptor Spindly and Dynactin. I identified a minimal domain of CENP-E that localises to these detachable structures and identified key residues essential for this localisation. By designing mutants, I showed that the kinetochore-targeting domain of CENP-E contains two kinetochore binding sites suggesting that two different pools of CENP-E are recruited to kinetochores. Finally, using siRNA, I showed that Dynein forms a second recruitment pathway of CENP-E to kinetochores. Overall, this work broadens the knowledge of kinetochore-microtubule attachment during mitosis in both yeast and humans. It provides key information that will allow future structural work on the Dam1 complex as well as future studies on the binding partners of CENP-E at kinetochores.
- Published
- 2021
- Full Text
- View/download PDF
11. P53 independent pathogenic mechanisms contribute to BubR1 microcephaly
- Author
-
Noelle A. Sterling, Bethany K. Terry, Julia M. McDonnell, and Seonhee Kim
- Subjects
BubR1 ,mitosis analysis ,neural progenitor ,p53 ,microcephaly ,cortical development ,Biology (General) ,QH301-705.5 - Abstract
The mosaic variegated aneuploidy (MVA)-associated gene Budding Uninhibited by Benzimidazole 1B (BUB1B) encodes BUBR1, a core member of the spindle assembly checkpoint complex that ensures kinetochore-spindle attachment for faithful chromosome segregation. BUB1B mutation in humans and its deletion in mice cause microcephaly. In the absence of BubR1 in mice, massive cell death reduces cortical cells during neurogenesis. However, the molecular and cellular mechanisms triggering cell death are unknown. In this study, we performed three-dimensional imaging analysis of mitotic BubR1-deficient neural progenitors in a murine model to show profound chromosomal segregation defects and structural abnormalities. Chromosomal defects and accompanying DNA damage result in P53 activation and apoptotic cell death in BubR1 mutants. To test whether the P53 cell death pathway is responsible for cortical cell loss, we co-deleted Trp53 in BubR1-deficient cortices. Remarkably, we discovered that residual apoptotic cell death remains in double mutants lacking P53, suggesting P53-independent apoptosis. Furthermore, the minimal rescue of cortical size and cortical neuron numbers in double mutant mice suggests the compelling extent of alternative death mechanisms in the absence of P53. This study demonstrates a potential pathogenic mechanism for microcephaly in MVA patients and uncovers the existence of powerful means of eliminating unfit cells even when the P53 death pathway is disabled.
- Published
- 2023
- Full Text
- View/download PDF
12. SIRT1 regulates mitotic catastrophe via autophagy and BubR1 signaling.
- Author
-
Zhao, Weiwei, Wang, Qing, Li, Le, Xie, Chengshen, Wu, Yequn, Gautam, Mayank, and Li, Lijia
- Abstract
Mitotic catastrophe (MC) is a suppressive mechanism that mediates the elimination of mitosis-deficient cells through apoptosis, necrosis or senescence after M phase block. SIRT1 is involved in the regulation of several cellular processes, including autophagy. However, the relationship between SIRT1 and MC has been largely obscure. Our study highlights that SIRT1 might be involved in the regulation of MC. We have shown that degradation of the SIRT1 protein via proteasome and lysosomal pathway was accompanied by MC induced via BMH-21. Overexpression of SIRT1 alleviated MC by decreasing the proportion of apoptotic and multinuclear cells induced by G2/M block and triggered autophagy whereas knockdown of SIRT1 aggravated MC and repressed autophagy. Furthermore, we found that serum starvation triggered autophagy evidently generated lower MC whereas siRNA of ATG5/7 suppressed autophagy leading to higher MC. ChIP analysis revealed that SIRT1 could bind to the promoter of BubR1, a component of spindle assembly checkpoint (SAC), to upregulate its expression. Overexpression of BubR1 decreased MC whereas knockdown of BubR1 increased it. These results reveal that SIRT1 regulates MC through autophagy and BubR1 signaling, and provide evidence for SIRT1, autophagy and BubR1 being the potential cancer therapeutic targets. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. BUBR1 as a Prognostic Biomarker in Canine Oral Squamous Cell Carcinoma.
- Author
-
Delgado, Leonor, Monteiro, Luís, Silva, Patrícia, Bousbaa, Hassan, Garcez, Fernanda, Silva, João, Brilhante-Simões, Paula, Pires, Isabel, and Prada, Justina
- Subjects
- *
SQUAMOUS cell carcinoma , *BIOMARKERS , *IMMUNOHISTOCHEMISTRY , *HUMAN carcinogenesis , *SURVIVAL rate - Abstract
Simple Summary: Spindle assembly checkpoint (SAC) includes several proteins that can be dysregulated contributing to oral carcinogenesis. We have investigated the role of some SAC components (BUBR1, BUB3 and SPINDLY proteins) in canine oral squamous cell carcinoma (OSCC) by immunohistochemical analysis of 60 canine OSCCs. We observed that all proteins were detected in almost all cases and with a high expression rate in some cases. Furthermore, we found an independent prognostic value for BUBR1, where high BUBR1 expression was associated with a lower survival rate of these canine patients. These results suggest a potential role of BUBR1 as a prognostic biomarker in canine OSCC and should motivate further studies aimed at the role of these SAC proteins not only as biomarkers but also as pharmacological targets in canine OSCC. Chromosomal instability (CIN) plays a key role in the carcinogenesis of several human cancers and can be related to the deregulation of core components of the spindle assembly checkpoint (SAC) including BUBR1 protein kinase. These proteins have been related to tumor development and poor survival rates in human patients with oral squamous cell carcinoma (OSCC). To investigate the expression of the SAC proteins BUBR1, BUB3 and SPINDLY and also Ki-67 in canine OSCC, we performed an immunohistochemical evaluation in 60 canine OSCCs and compared them with clinical and pathological variables. BUBR1, Ki-67, BUB3 and SPINDLY protein expressions were detected in all cases and classified as with a high-expression extent score in 31 (51.7%) cases for BUBR1, 33 (58.9%) cases for BUB3 and 28 (50.9%) cases for SPINDLY. Ki-67 high expression was observed in 14 (25%) cases. An independent prognostic value for BUBR1 was found, where high BUBR1 expression was associated with lower survival (p = 0.012). These results indicate that BUBR1 expression is an independent prognostic factor in these tumors, suggesting the potential use for clinical applications as a prognostic biomarker and also as a pharmacological target in canine OSCC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Spatial separation of phosphatase and kinase activity within the Bub complex is required for proper mitosis.
- Author
-
Wang, Lei, Kruse, Thomas, López-Méndez, Blanca, Zhang, Yuqing, Song, Chunlin, Zhu, Lei, Li, Bing, Fang, Jing, Lu, Zhimin, Nilsson, Jakob, and Zhang, Gang
- Abstract
The Bub1 and BubR1 kinetochore proteins support proper chromosome segregation and mitotic checkpoint activity. Bub1 and BubR1 are paralogs with Bub1 being a kinase, while BubR1 localizes the PP2A-B56 protein phosphatase to kinetochores in humans. Whether this spatial separation of kinase and phosphatase activity is important is unclear as some organisms integrate both activities into one Bub protein. Here, we engineer human Bub1 and BubR1 proteins integrating kinase and phosphatase activities into one protein and show that these do not support normal mitotic progression. A Bub1–PP2A-B56 complex can support chromosome alignment but results in impairment of the checkpoint due to dephosphorylation of the Mad1 binding site in Bub1. Furthermore, a chimeric BubR1 protein containing the Bub1 kinase domain induces delocalized H2ApT120 phosphorylation, resulting in the reduction of centromeric hSgo2 and chromosome segregation errors. Collectively, these results argue that the spatial separation of kinase and phosphatase activities within the Bub complex is required for balancing its functions in the checkpoint and chromosome alignment. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
15. Hyper-active RAS/MAPK introduces cancer-specific mitotic vulnerabilities.
- Author
-
Herman, Jacob A., Romain, Romario R., Hoellerbauer, Pia, Shirnekhi, Hazheen K., King, David C., DeLuca, Keith F., Nishimura, Erin Osborne, Paddison, Patrick J., and DeLuca, Jennifer G.
- Subjects
- *
MITOGEN-activated protein kinases , *CHROMOSOME segregation , *KINETOCHORE , *ANEUPLOIDY , *INTEGERS - Abstract
Aneuploidy, the incorrect number of whole chromosomes, is a common feature of tumors that contributes to their initiation and evolution. Preventing aneuploidy requires properly functioning kinetochores, which are large protein complexes assembled on centromeric DNA that link mitotic chromosomes to dynamic spindle microtubules and facilitate chromosome segregation. The kinetochore leverages at least two mechanisms to prevent aneuploidy: error correction and the spindle assembly checkpoint (SAC). BubR1, a factor involved in both processes, was identified as a cancer dependency and therapeutic target in multiple tumor types; however, it remains unclear what specific oncogenic pressures drive this enhanced dependency on BubR1 and whether it arises from BubR1’s regulation of the SAC or error-correction pathways. Here, we use a genetically controlled transformation model and glioblastoma tumor isolates to show that constitutive signaling by RAS or MAPK is necessary for cancer-specific BubR1 vulnerability. TheMAPK pathway enzymatically hyperstimulates a network of kinetochore kinases that compromises chromosome segregation, rendering cells more dependent on two BubR1 activities: counteracting excessive kinetochore–microtubule turnover for error correction and maintaining the SAC. This work expands our understanding of how chromosome segregation adapts to different cellular states and reveals an oncogenic trigger of a cancer-specific defect. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
16. Physiological relevance of post-translational regulation of the spindle assembly checkpoint protein BubR1
- Author
-
Celia R. Bloom and Brian J. North
- Subjects
Post-translational modifications ,BubR1 ,Mitosis ,Acetylation ,Phosphorylation ,Ubiquitination ,Biotechnology ,TP248.13-248.65 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Abstract BubR1 is an essential component of the spindle assembly checkpoint (SAC) during mitosis where it functions to prevent anaphase onset to ensure proper chromosome alignment and kinetochore-microtubule attachment. Loss or mutation of BubR1 results in aneuploidy that precedes various potential pathologies, including cancer and mosaic variegated aneuploidy (MVA). BubR1 is also progressively downregulated with age and has been shown to be directly involved in the aging process through suppression of cellular senescence. Post-translational modifications, including but not limited to phosphorylation, acetylation, and ubiquitination, play a critical role in the temporal and spatial regulation of BubR1 function. In this review, we discuss the currently characterized post-translational modifications to BubR1, the enzymes involved, and the biological consequences to BubR1 functionality and implications in diseases associated with BubR1. Understanding the molecular mechanisms promoting these modifications and their roles in regulating BubR1 is important for our current understanding and future studies of BubR1 in maintaining genomic integrity as well as in aging and cancer.
- Published
- 2021
- Full Text
- View/download PDF
17. Molecular contribution to embryonic aneuploidy and karyotypic complexity in initial cleavage divisions of mammalian development.
- Author
-
Brooks, Kelsey E., Daughtry, Brittany L., Davis, Brett, Yan, Melissa Y., Fei, Suzanne S., Shepherd, Selma, Carbone, Lucia, and Chavez, Shawn L.
- Subjects
- *
ANEUPLOIDY , *CELL anatomy , *REPRODUCTIVE technology , *NUCLEOLUS , *CHROMOSOME segregation , *NUCLEAR structure , *CELL division , *CENTROMERE - Abstract
Embryonic aneuploidy is highly complex, often leading to developmental arrest, implantation failure or spontaneous miscarriage in both natural and assisted reproduction. Despite our knowledge of mitotic mis-segregation in somatic cells, the molecular pathways regulating chromosome fidelity during the error-prone cleavage-stage of mammalian embryogenesis remain largely undefined. Using bovine embryos and live-cell fluorescent imaging, we observed frequent micro-/multi-nucleation of mis-segregated chromosomes in initial mitotic divisions that underwent unilateral inheritance, re-fused with the primary nucleus or formed a chromatin bridge with neighboring cells. A correlation between a lack of syngamy, multipolar divisions and asymmetric genome partitioning was also revealed, and single-cell DNA-seq showed propagation of primarily non-reciprocal mitotic errors. Depletion of the mitotic checkpoint protein BUB1B (also known as BUBR1) resulted in similarly abnormal nuclear structures and cell divisions, as well as chaotic aneuploidy and dysregulation of the kinase-substrate network that mediates mitotic progression, all before zygotic genome activation. This demonstrates that embryonic micronuclei sustain multiple fates, provides an explanation for blastomeres with uniparental origins, and substantiates defective checkpoints and likely othermaternally derived factors as major contributors to the karyotypic complexity afflicting mammalian preimplantation development. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. BUBR1
- Author
-
Braga, Luciano Gama, Elowe, Sabine, and Choi, Sangdun, editor
- Published
- 2018
- Full Text
- View/download PDF
19. BUBR1 as a Prognostic Biomarker in Canine Oral Squamous Cell Carcinoma
- Author
-
Leonor Delgado, Luís Monteiro, Patrícia Silva, Hassan Bousbaa, Fernanda Garcez, João Silva, Paula Brilhante-Simões, Isabel Pires, and Justina Prada
- Subjects
oral cancer ,BUBR1 ,BUB3 ,SPINDLY ,Ki-67 ,immunohistochemistry ,Veterinary medicine ,SF600-1100 ,Zoology ,QL1-991 - Abstract
Chromosomal instability (CIN) plays a key role in the carcinogenesis of several human cancers and can be related to the deregulation of core components of the spindle assembly checkpoint (SAC) including BUBR1 protein kinase. These proteins have been related to tumor development and poor survival rates in human patients with oral squamous cell carcinoma (OSCC). To investigate the expression of the SAC proteins BUBR1, BUB3 and SPINDLY and also Ki-67 in canine OSCC, we performed an immunohistochemical evaluation in 60 canine OSCCs and compared them with clinical and pathological variables. BUBR1, Ki-67, BUB3 and SPINDLY protein expressions were detected in all cases and classified as with a high-expression extent score in 31 (51.7%) cases for BUBR1, 33 (58.9%) cases for BUB3 and 28 (50.9%) cases for SPINDLY. Ki-67 high expression was observed in 14 (25%) cases. An independent prognostic value for BUBR1 was found, where high BUBR1 expression was associated with lower survival (p = 0.012). These results indicate that BUBR1 expression is an independent prognostic factor in these tumors, suggesting the potential use for clinical applications as a prognostic biomarker and also as a pharmacological target in canine OSCC.
- Published
- 2022
- Full Text
- View/download PDF
20. Adventures of the undead at kinetochores
- Author
-
Sabine Elowe
- Subjects
mitosis ,kinetochore ,bubr1 ,pseudokinase ,pp2a ,b56 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
At the metaphase-to-anaphase transition, phosphatase activity feeds back to reverse early mitotic phosphorylation events. Our recent work indicates that the pseudokinase domain of the spindle checkpoint protein BUB1 (Budding Uninhibited by Benzimidazoles 1) mitotic checkpoint serine/threonine kinase B (BUB1B, BUBR1) maintains kinase–phosphatase balance at the outer kinetochore during mitotic exit.
- Published
- 2021
- Full Text
- View/download PDF
21. Physiological relevance of post-translational regulation of the spindle assembly checkpoint protein BubR1.
- Author
-
Bloom, Celia R. and North, Brian J.
- Subjects
- *
POST-translational modification , *CELLULAR aging , *MICROTUBULES , *UBIQUITINATION , *PROTEINS , *ANEUPLOIDY , *ANAPHASE , *CHROMOSOMES - Abstract
BubR1 is an essential component of the spindle assembly checkpoint (SAC) during mitosis where it functions to prevent anaphase onset to ensure proper chromosome alignment and kinetochore-microtubule attachment. Loss or mutation of BubR1 results in aneuploidy that precedes various potential pathologies, including cancer and mosaic variegated aneuploidy (MVA). BubR1 is also progressively downregulated with age and has been shown to be directly involved in the aging process through suppression of cellular senescence. Post-translational modifications, including but not limited to phosphorylation, acetylation, and ubiquitination, play a critical role in the temporal and spatial regulation of BubR1 function. In this review, we discuss the currently characterized post-translational modifications to BubR1, the enzymes involved, and the biological consequences to BubR1 functionality and implications in diseases associated with BubR1. Understanding the molecular mechanisms promoting these modifications and their roles in regulating BubR1 is important for our current understanding and future studies of BubR1 in maintaining genomic integrity as well as in aging and cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
22. Expression of spindle assembly checkpoint proteins BubR1 and Mad2 expression as potential biomarkers of malignant transformation of oral leukoplakia: an observational cohort study.
- Author
-
Monteiro, Luís, Silva, Patrícia, Delgado, Leonor, Amaral, Barbas, Garcês, Fernanda, Salazar, Filomena, Júlio Pacheco, José, Lopes, Carlos, Bousbaa, Hassan, and Warnakulasuriya, Saman
- Subjects
ORAL leukoplakia ,CHROMOSOME segregation ,UNIVARIATE analysis ,LEUKOPLAKIA ,COHORT analysis ,MULTIVARIATE analysis ,ORAL mucosa - Abstract
Background: The Spindle Assembly Checkpoint (SAC) is a surveillance mechanism essential to ensure the accuracy of chromosome segregation during mitosis. Our aim was to evaluate the expression of SAC proteins in oral carcinogenesis, and to assess their potential in predicting malignant transformation of oral leukoplakia. Material and Methods: We analysed the immunoexpression of BubR1, Mad2, Bub3, and Spindly proteins in 64 oral biopsies from 52 oral leukoplakias and 12 normal tissues. Univariate and multivariate analysis were performed to evaluate predictive factors for malignant transformation (MT). Results: We observed that BubR1 and Mad2 were more highly expressed in high dysplasia grade lesions than in low grade or normal tissues (P<0.05). High expression of Spindly was significantly correlated with a high Ki-67 score (P=0.004). Six (11.5%) oral leukoplakias underwent malignant transformation. In univariate analysis, the binary dysplasia grade (high grade) (P<0.001) was associated with a higher risk of malignant transformation as well as high BubR1 (P<0.001) and high Mad2 (P=0.013) expression. In multivariate analysis, high expression of BubR1 and Mad2 when combined showed an increased risk for malignant transformation (P=0.013; HR of 4.6, 95% CI of 1.4-15.1). Conclusions: Our findings reveal that BubR1 and Mad2 were associated with an increased risk for malignant transformation independently of histological grade and could be potential and useful predictive risk markers of malignant transformation in oral leukoplakias. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
23. BUBR1 Pseudokinase Domain Promotes Kinetochore PP2A-B56 Recruitment, Spindle Checkpoint Silencing, and Chromosome Alignment
- Author
-
Luciano Gama Braga, Angel F. Cisneros, Michelle M. Mathieu, Maxime Clerc, Pauline Garcia, Baptiste Lottin, Chantal Garand, Philippe Thebault, Christian R. Landry, and Sabine Elowe
- Subjects
BUBR1 ,pseudokinase ,PP2A ,B56 ,Kinetochore ,Spindle checkpoint ,Biology (General) ,QH301-705.5 - Abstract
Summary: The balance of phospho-signaling at the outer kinetochore is critical for forming accurate attachments between kinetochores and the mitotic spindle and timely exit from mitosis. A major player in determining this balance is the PP2A-B56 phosphatase, which is recruited to the kinase attachment regulatory domain (KARD) of budding uninhibited by benzimidazole 1-related 1 (BUBR1) in a phospho-dependent manner. This unleashes a rapid, switch-like phosphatase relay that reverses mitotic phosphorylation at the kinetochore, extinguishing the checkpoint and promoting anaphase. Here, we demonstrate that the C-terminal pseudokinase domain of human BUBR1 is required to promote KARD phosphorylation. Mutation or removal of the pseudokinase domain results in decreased PP2A-B56 recruitment to the outer kinetochore attenuated checkpoint silencing and errors in chromosome alignment as a result of imbalance in Aurora B activity. Our data, therefore, elucidate a function for the BUBR1 pseudokinase domain in ensuring accurate and timely exit from mitosis.
- Published
- 2020
- Full Text
- View/download PDF
24. BUB1 Is Essential for the Viability of Human Cells in which the Spindle Assembly Checkpoint Is Compromised
- Author
-
Jonne A. Raaijmakers, Roy G.H.P. van Heesbeen, Vincent A. Blomen, Louise M.E. Janssen, Ferdy van Diemen, Thijn R. Brummelkamp, and René H. Medema
- Subjects
spindle assembly checkpoint ,BUB1 ,BUBR1 ,MAD1 ,MAD2 ,chromosome alignment ,Biology (General) ,QH301-705.5 - Abstract
Summary: The spindle assembly checkpoint (SAC) ensures faithful segregation of chromosomes. Although most mammalian cell types depend on the SAC for viability, we found that human HAP1 cells can grow SAC independently. We generated MAD1- and MAD2-deficient cells and mutagenized them to identify synthetic lethal interactions, revealing that chromosome congression factors become essential upon SAC deficiency. Besides expected hits, we also found that BUB1 becomes essential in SAC-deficient cells. We found that the BUB1 C terminus regulates alignment as well as recruitment of CENPF. Second, we found that BUBR1 was not essential in SAC-deficient HAP1 cells. We confirmed that BUBR1 does not regulate chromosome alignment in HAP1 cells and that BUB1 does not regulate chromosome alignment through BUBR1. Taken together, our data resolve some long-standing questions about the interplay between BUB1 and BUBR1 and their respective roles in the SAC and chromosome alignment.
- Published
- 2018
- Full Text
- View/download PDF
25. Downregulation of Mad2 and BubR1 increase the malignant potential and nocodazole resistance by compromising spindle assembly checkpoint signaling pathway in cervical carcinogenesis.
- Author
-
Wang, Li, Wang, Jian, Jin, Yubiao, Zheng, Jun, Yang, Yongbin, and Xi, Xiaowei
- Subjects
- *
PROTEIN metabolism , *CARCINOGENESIS , *ANTINEOPLASTIC agents , *APOPTOSIS , *CELL cycle , *CELL migration , *CELLULAR signal transduction , *CERVICAL cancer , *FLOW cytometry , *GENE expression , *IMMUNOHISTOCHEMISTRY , *MICROBIOLOGICAL assay , *RNA , *SQUAMOUS cell carcinoma , *STAINS & staining (Microscopy) , *CELL survival , *PHARMACODYNAMICS - Abstract
Aim: To explore the involvement of Mad2 and BubR1 in cervical carcinogenesis. Methods: The expressions of Mad2 and BubR1 in tissues of high‐grade squamous intraepithelial lesions (HSIL), low‐grade squamous intraepithelial lesions (LSIL) and chronic cervicitis were analyzed immunohistochemistrily and compared with those of p16INK4A. PEGFP‐Mad2 and pEGFP‐BubR1 were transfected into SiHa cells to overexpress Mad2 and BubR1 and Si‐RNAs to knockdown. Cell viability was measured by cell counting kit‐8 (CCK‐8) assay. Migration and invasion capabilities were detected by Transwell. Propidium iodide staining with flow cytometry was used for cell cycle analysis and apoptosis was detected using Annexin V/7‐AAD staining after nocodazole treatment. Results: The expression of Mad2 was significantly lower in HSIL than those in chronic cervicitis and LSIL, however, the expression of BubR1 showed no significant differences. To detect HSIL in cervical lesions, Mad2 had a sensitivity of 88.44% and a specificity of 87.23%, Mad2 was less sensitive and more specific than p16INK4a. In SiHa cells, knockdown of Mad2 and BubR1 increased cell growth, reinforced invasion capacity and migration potency, inhibited apoptosis and decreased G2‐phase distribution after nocodazole treatment. Oppositely, the overexpression strategies made cells show decreased malignant behaviors, raised apoptosis and increased G2‐phase distribution. Conclusion: Mad2 negativity was specific to identify HSIL immunohistochemistrily. Downregulation of Mad2 and BubR1 increase the malignant behavior and nocodazole resistance of SiHa cells via causing spindle assembly checkpoint defect. This mechanism may contribute to cervical carcinogenesis and resistance to microtubule‐targeting drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
26. BubR1 and cyclin B1 immunoexpression in pleomorphic adenoma and polymorphous adenocarcinoma of minor salivary glands.
- Author
-
Cavalcante, Israel Leal, Silva Barros, Caio César da, Colares, Débora Frota, Cruz, Vitória Maria Sousa, de Andrade, Bruno Augusto Benevenuto, Nonaka, Cassiano Francisco Weege, Rabenhorst, Silvia Helena Barem, and Cavalcante, Roberta Barroso
- Subjects
- *
PLEOMORPHIC adenoma , *SALIVARY glands , *CYCLINS , *CELL cycle proteins , *IMMUNOHISTOCHEMISTRY , *ADENOCARCINOMA - Abstract
The immunoexpression of BubR1 and cyclin B1 in pleomorphic adenoma (PA) and polymorphic adenocarcinoma (PAC) in minor salivary glands is poorly studied. Thus, a retrospective and observational study was performed to provide a better understanding of the role and immunopositivity patterns of these proteins in these lesions. Sixteen cases of PA and 16 cases of PAC were selected. Parenchyma cells were submitted to quantitative immunohistochemical analysis through the labeling index. Cytoplasmic immunoexpression of BubR1 was observed in neoplastic cells from all analyzed PA and PAC cases. All PA cases and 93.7% of PAC exhibited nuclear immunoexpression of BubR1. Higher cytoplasmic and nuclear immunoexpression of BubR1 was observed in PAC (p = 0.001 and p = 0.122, respectively). Cytoplasmic immunoexpression of cyclin B1 was observed in all cases of PA and PAC, with a higher labeling index in the latter (p < 0.001). There was a significant positive correlation between nuclear and cytoplasmic BubR1 immunoexpressions (p < 0.001) in PA and a significant negative correlation between BubR1 and cyclin B1 cytoplasmic immunoexpressions (p = 0.014) in PAC. The higher cytoplasmic and nuclear immunoexpression of BubR1 in PACs suggests the continuous maintenance of neoplastic cells in the cell cycle and migration. Higher immunoexpression of cyclin B1 supports this lesion's enhanced proliferative and migration ability. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Centromere-associated protein-E is essential for the mammalian mitotic checkpoint to prevent aneuploidy due to single chromosome loss
- Author
-
Weaver, Beth AA, Bonday, Zahid Q, Putkey, Frances R, Kops, Geert JPL, Silk, Alain D, and Cleveland, Don W
- Subjects
Genetics ,1.1 Normal biological development and functioning ,Underpinning research ,Aneuploidy ,Animals ,Cell Cycle Proteins ,Centromere ,Chromosomal Proteins ,Non-Histone ,Fibroblasts ,HeLa Cells ,Humans ,Integrases ,Kinetochores ,Mice ,Mitosis ,Protein Kinases ,Protein Serine-Threonine Kinases ,Sequence Analysis ,DNA ,Viral Proteins ,kinetochore ,mitosis ,cell cycle ,LENP-E ,BubR1 ,Hela Cells ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Centromere-associated protein-E (CENP-E) is an essential mitotic kinesin that is required for efficient, stable microtubule capture at kinetochores. It also directly binds to BubR1, a kinetochore-associated kinase implicated in the mitotic checkpoint, the major cell cycle control pathway in which unattached kinetochores prevent anaphase onset. Here, we show that single unattached kinetochores depleted of CENP-E cannot block entry into anaphase, resulting in aneuploidy in 25% of divisions in primary mouse fibroblasts in vitro and in 95% of regenerating hepatocytes in vivo. Without CENP-E, diminished levels of BubR1 are recruited to kinetochores and BubR1 kinase activity remains at basal levels. CENP-E binds to and directly stimulates the kinase activity of purified BubR1 in vitro. Thus, CENP-E is required for enhancing recruitment of its binding partner BubR1 to each unattached kinetochore and for stimulating BubR1 kinase activity, implicating it as an essential amplifier of a basal mitotic checkpoint signal.
- Published
- 2003
28. BubR1 controls starvation-induced lipolysis via IMD signaling pathway in Drosophila .
- Author
-
Liu M, Yang S, Yang J, Feng P, Luo F, Zhang Q, Yang L, and Jiang H
- Subjects
- Animals, Lipolysis, Signal Transduction, Lipids, Cell Cycle Proteins metabolism, Drosophila genetics, Drosophila Proteins metabolism
- Abstract
Lipolysis, the key process releasing fat acids to generate energy in adipose tissues, correlates with starvation resistance. Nevertheless, its detail mechanisms remain elusive. BubR1, an essential mitotic regulator, ensures proper chromosome alignment and segregation during mitosis, but its physiological functions are largely unknown. Here, we use Drosophila adult fat body, the major lipid storage organ, to study the functions of BubR1 in lipolysis. We show that both whole body- and fat body-specific BubR1 depletions increase lipid degradation and shorten the lifespan under fasting but not feeding. Relish, the conserved regulator of IMD signaling pathway, acts as the downstream target of BubR1 to control the expression level of Bmm and modulate the lipolysis upon fasting. Thus, our study reveals new functions of BubR1 in starvation-induced lipolysis and provides new insights into the molecular mechanisms of lipolysis mediated by IMD signaling pathway.
- Published
- 2024
- Full Text
- View/download PDF
29. Rhabdomyosarcoma with premature chromatid separation–mosaic variegated aneuploidy syndrome: Reduced‐intensity chemotherapy.
- Author
-
Nishida, Yoshinobu, Usami, Ikuya, Nishitani‐Isa, Masahiko, Maihara, Toshiro, and Hiraumi, Yoshimi
- Subjects
- *
ANEUPLOIDY , *ANTINEOPLASTIC agents , *CANCER chemotherapy , *CHROMOSOME abnormalities , *MAGNETIC resonance imaging , *MOSAICISM , *QUALITY of life , *RHABDOMYOSARCOMA , *GENETIC testing - Abstract
The article presents a case study of a female patient diagnosed with PCS(MVA) syndrome and treated with reduced-dose chemotherapy for rhabdomyosarcoma. Topics include how prognosis and quality of life can be improved after a diagnosis of PCS(MVA) syndrome if the malignant tumor is found early and proper intervention is initiated.
- Published
- 2019
- Full Text
- View/download PDF
30. Overexpression of Fbxo6 inactivates spindle checkpoint by interacting with Mad2 and BubR1.
- Author
-
Xu, Han-Zhang, Wang, Zhuo-Qun, Shan, Hui-Zhuang, Zhou, Li, Yang, Li, Lei, Hu, Liu, Bin, and Wu, Ying-Li
- Abstract
The spindle assembly checkpoint prevents chromosome mis-segregation during mitosis by delaying sister chromatid separation. Several F-box protein members play critical roles in maintaining genome stability and regulating cell cycle progress via ubiquitin-mediated protein degradation. Here, we showed that Fbxo6 critically regulated spindle checkpoint and chromosome segregation. Fbxo6 was phosphorylated during mitosis. Overexpression of Fbxo6 lead to faster exit from nocodazole-induced mitosis arrest through premature sister chromatid separation. Moreover, we found substantially more binuclear and multilobed nuclei cells accompanied with impaired cell viability in Fbxo6-overexpressed HeLa cells. Mechanistically, Fbxo6 interacted with spindle checkpoint proteins including Mad2 and BubR1 leading to the premature exit from mitosis. Overall, we revealed a novel role of Fbxo6 in regulating spindle checkpoint, which may shed light on the regulation of genome instability of cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
31. The mitotic checkpoint complex (MCC): looking back and forth after 15 years
- Author
-
Song-Tao Liu and Hang Zhang
- Subjects
mitotic checkpoint ,mitotic checkpoint complex ,anaphase promoting complex/cyclosome ,ubiquitin ligase ,BUBR1 ,MAD2 ,CDC20 ,chromosome segregation ,Biology (General) ,QH301-705.5 - Abstract
The mitotic checkpoint is a specialized signal transduction pathway that contributes to the fidelity of chromosome segregation. The signaling of the checkpoint originates from defective kinetochore-microtubule interactions and leads to formation of the mitotic checkpoint complex (MCC), a highly potent inhibitor of the Anaphase Promoting Complex/Cyclosome (APC/C)—the E3 ubiquitin ligase essential for anaphase onset. Many important questions concerning the MCC and its interaction with APC/C have been intensively investigated and debated in the past 15 years, such as the exact composition of the MCC, how it is assembled during a cell cycle, how it inhibits APC/C, and how the MCC is disassembled to allow APC/C activation. These efforts have culminated in recently reported structure models for human MCC:APC/C supra-complexes at near-atomic resolution that shed light on multiple aspects of the mitotic checkpoint mechanisms. However, confusing statements regarding the MCC are still scattered in the literature, making it difficult for students and scientists alike to obtain a clear picture of MCC composition, structure, function and dynamics. This review will comb through some of the most popular concepts or misconceptions about the MCC, discuss our current understandings, present a synthesized model on regulation of CDC20 ubiquitination, and suggest a few future endeavors and cautions for next phase of MCC research.
- Published
- 2016
- Full Text
- View/download PDF
32. Crystal structure of a PP2A B56-BubR1 complex and its implications for PP2A substrate recruitment and localization
- Author
-
Jiao Wang, Zhizhi Wang, Tingting Yu, Huan Yang, David M. Virshup, Geert J. P. L. Kops, Sang Hyun Lee, Weihong Zhou, Xin Li, Wenqing Xu, and Zihe Rao
- Subjects
PP2A ,BubR1 ,kinetochore ,cellular targeting ,substrate recruitment ,Cytology ,QH573-671 ,Animal biochemistry ,QP501-801 - Abstract
ABSTRACT Protein phosphatase 2A (PP2A) accounts for the majority of total Ser/Thr phosphatase activities in most cell types and regulates many biological processes. PP2A holoenzymes contain a scaffold A subunit, a catalytic C subunit, and one of the regulatory/targeting B subunits. How the B subunit controls PP2A localization and substrate specificity, which is a crucial aspect of PP2A regulation, remains poorly understood. The kinetochore is a critical site for PP2A functioning, where PP2A orchestrates chromosome segregation through its interactions with BubR1. The PP2A-BubR1 interaction plays important roles in both spindle checkpoint silencing and stable microtubule-kinetochore attachment. Here we present the crystal structure of a PP2A B56-BubR1 complex, which demonstrates that a conserved BubR1 LxxIxE motif binds to the concave side of the B56 pseudo-HEAT repeats. The BubR1 motif binds to a groove formed between B56 HEAT repeats 3 and 4, which is quite distant from the B56 binding surface for PP2A catalytic C subunit and thus is unlikely to affect PP2A activity. In addition, the BubR1 binding site on B56 is far from the B56 binding site of shugoshin, another kinetochore PP2A-binding protein, and thus BubR1 and shugoshin can potentially interact with PP2A-B56 simultaneously. Our structural and biochemical analysis indicates that other proteins with the LxxIxE motif may also bind to the same PP2A B56 surface. Thus, our structure of the PP2A B56-BubR1 complex provides important insights into how the B56 subunit directs the recruitment of PP2A to specific targets.
- Published
- 2016
- Full Text
- View/download PDF
33. Haplo-insufficiency of both BubR1 and SGO1 accelerates cellular senescence
- Author
-
Sung-Hyun Park, Steve Xie, Chinthalapally V. Rao, and Wei Dai
- Subjects
BubR1 ,SGO1 ,Knockout mice ,MEFs ,Senescence ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Spindle assembly checkpoint components BubR1 and Sgo1 play a key role in the maintenance of chromosomal instability during cell division. These proteins function to block the anaphase entry until all condensed chromosomes have been attached by the microtubules emanating from both spindle poles. Haplo-insufficiency of either BubR1 or SGO1 results in enhanced chromosomal instability and tumor development in the intestine. Recent studies show that spindle checkpoint proteins also have a role in slowing down the ageing process. Therefore, we want to study whether haplo-insufficiency of both BubR1 and SGO1 accelerates cellular senescence in mice. Methods We took advantage of the availability of BubR1 and SGO1 knockout mice and generated primary murine embryonic fibroblasts (MEFs) with mutations in either BubR1, SGO1, or both and analyzed cellular senescence of the MEFs of various genetic backgrounds. Results We observed that BubR1 +/− SGO +/− MEFs had an accelerated cellular senescence characterized by morphological changes and expressed senescence-associated β-galactosidase. In addition, compared with wild-type MEFs or MEFs with a single gene deficiency, BubR1 +/− SGO1 +/− MEFs expressed enhanced levels of p21 but not p16. Conclusions Taken together, our observations suggest that combined deficiency of BubR1 and Sgo1 accelerates cellular senescence.
- Published
- 2016
- Full Text
- View/download PDF
34. Discovery of Novel Agents on Spindle Assembly Checkpoint to Sensitize Vinorelbine-Induced Mitotic Cell Death against Human Non-Small Cell Lung Cancers
- Author
-
Ya-Ching Chang, Yu-Ling Tseng, Wohn-Jenn Leu, Chi-Min Du, Yi-Huei Jiang, Lih-Ching Hsu, Jui-Ling Hsu, Duen-Ren Hou, and Jih-Hwa Guh
- Subjects
Novel sildenafil mimetic ,vinorelbine ,spindle assembly checkpoint ,apoptotic synergism ,BUBR1 ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Non-small cell lung cancer (NSCLC) accounts about 80% of all lung cancers. More than two-thirds of NSCLC patients have inoperable, locally advanced or metastatic tumors. Non-toxic agents that synergistically potentiate cancer-killing activities of chemotherapeutic drugs are in high demand. YL-9 was a novel and non-cytotoxic compound with the structure related to sildenafil but showing much less activity against phosphodiesterase type 5 (PDE5). NCI-H460, an NSCLC cell line with low PDE5 expression, was used as the cell model. YL-9 synergistically potentiated vinorelbine-induced anti-proliferative and apoptotic effects in NCI-H460 cells. Vinorelbine induced tubulin acetylation and Bub1-related kinase (BUBR1) phosphorylation, a necessary component in spindle assembly checkpoint. These effects, as well as BUBR1 cleavage, were substantially enhanced in co-treatment with YL-9. Several mitotic arrest signals were enhanced under combinatory treatment of vinorelbine and YL-9, including an increase of mitotic spindle abnormalities, increased cyclin B1 expression, B-cell lymphoma 2 (Bcl-2) phosphorylation and increased phosphoproteins. Moreover, YL-9 also displayed synergistic activity in combining with vinorelbine to induce apoptosis in A549 cells which express PDE5. In conclusion. the data suggest that YL-9 is a novel agent that synergistically amplifies vinorelbine-induced NSCLC apoptosis through activation of spindle assembly checkpoint and increased mitotic arrest of the cell cycle. YL-9 shows the potential for further development in combinatory treatment against NSCLC.
- Published
- 2020
- Full Text
- View/download PDF
35. BubR1 Insufficiency Impairs Affective Behavior and Memory Function in Mice
- Author
-
Chang Hoon Cho, Zhongxi Yang, Ki Hyun Yoo, Alfredo Oliveros, and Mi Hyeon Jang
- Subjects
BubR1 ,Aging ,Hippocampus ,Memory ,Emotion ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Purpose Although aging causes functional declines in cognition, the molecular mechanism underlying these declines remains largely unknown. Recently, the spindle checkpoint kinase budding uninhibited by benzimidazole-related 1 (BubR1) has emerged as a key determinant for age-related pathology in various tissues including brain. However, the neurobehavioral impact of BubR1 has not been explored. In this study, we investigated the role of BubR1 in behavioral function. Methods To investigate the neurobiological functions of BubR1 in vivo, we utilized transgenic mice harboring BubR1 hypomorphic alleles (BubR1H/H mice), which produce low amounts of BubR1 protein, as well as mice that have specific knockdown of BubR1 in the adult dentate gyrus. To assess anxiety-like behavior, the above groups were subjected to the elevated plus maze and the light-dark test, in addition to utilizing the tail-suspension and forced-swim test to determine depression-like behavior. We used novel object recognition to test for memory-related function. Results We found that BubR1H/H mice display several behavioral deficits when compared to wild-type littermates, including increased anxiety in the elevated-plus maze test, depression-like behavior in the tail suspension test, as well as impaired memory function in the novel object recognition test. Similar to BubR1H/H mice, knockdown of BubR1 within the adult dentate gyrus led to increased anxiety-like behavior as well as depression-like behavior, and impaired memory function. Conclusions Our study demonstrates a requirement of BubR1 in maintaining proper affective and memory-related behavioral function. These results suggest that a decline in BubR1 levels with advanced age may be a crucial contributor to age-related hippocampal dysfunction.
- Published
- 2018
- Full Text
- View/download PDF
36. BubR1 Acts as a Promoter in Cellular Motility of Human Oral Squamous Cancer Cells through Regulating MMP-2 and MMP-9
- Author
-
Chou-Kit Chou, Chang-Yi Wu, Jeff Yi-Fu Chen, Ming-Chong Ng, Hui-Min David Wang, Jen-Hao Chen, Shyng-Shiou F. Yuan, Eing-Mei Tsai, Jan-Gowth Chang, and Chien-Chih Chiu
- Subjects
BubR1 ,spindle assembly checkpoint ,oral squamous cancer cell ,migration ,metastasis ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
BubR1 is a critical component of spindle assembly checkpoint, ensuring proper chromatin segregation during mitosis. Recent studies showed that BubR1 was overexpressed in many cancer cells, including oral squamous cell carcinomas (OSCC). However, the effect of BubR1 on metastasis of OSCC remains unclear. This study aimed to unravel the role of BubR1 in the progression of OSCC and confirm the expression of BubR1 in a panel of malignant OSCC cell lines with different invasive abilities. The results of quantitative real-time PCR showed that the mRNA level of BubR1 was markedly increased in four OSCC cell lines, Ca9-22, HSC3, SCC9 and Cal-27 cells, compared to two normal cells, normal human oral keratinocytes (HOK) and human gingival fibroblasts (HGF). Moreover, the expression of BubR1 in these four OSCC cell lines was positively correlated with their motility. Immunofluorescence revealed that BubR1 was mostly localized in the cytosol of human gingival carcinoma Ca9-22 cells. BubR1 knockdown significantly decreased cellular invasion but slightly affect cellular proliferation on both Ca9-22 and Cal-27 cells. Consistently, the activities of metastasis-associated metalloproteinases MMP-2 and MMP-9 were attenuated in BubR1 knockdown Ca9-22 cells, suggesting the role of BubR1 in promotion of OSCC migration. Our present study defines an alternative pathway in promoting metastasis of OSCC cells, and the expression of BubR1 could be a prognostic index in OSCC patients.
- Published
- 2015
- Full Text
- View/download PDF
37. P53 independent pathogenic mechanisms contribute to BubR1 microcephaly.
- Author
-
Sterling NA, Terry BK, McDonnell JM, and Kim S
- Abstract
The mosaic variegated aneuploidy (MVA)-associated gene Budding Uninhibited by Benzimidazole 1B ( BUB1B ) encodes BUBR1, a core member of the spindle assembly checkpoint complex that ensures kinetochore-spindle attachment for faithful chromosome segregation. BUB1B mutation in humans and its deletion in mice cause microcephaly. In the absence of BubR1 in mice, massive cell death reduces cortical cells during neurogenesis. However, the molecular and cellular mechanisms triggering cell death are unknown. In this study, we performed three-dimensional imaging analysis of mitotic BubR1-deficient neural progenitors in a murine model to show profound chromosomal segregation defects and structural abnormalities. Chromosomal defects and accompanying DNA damage result in P53 activation and apoptotic cell death in BubR1 mutants. To test whether the P53 cell death pathway is responsible for cortical cell loss, we co-deleted Trp53 in BubR1-deficient cortices. Remarkably, we discovered that residual apoptotic cell death remains in double mutants lacking P53, suggesting P53-independent apoptosis. Furthermore, the minimal rescue of cortical size and cortical neuron numbers in double mutant mice suggests the compelling extent of alternative death mechanisms in the absence of P53. This study demonstrates a potential pathogenic mechanism for microcephaly in MVA patients and uncovers the existence of powerful means of eliminating unfit cells even when the P53 death pathway is disabled., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Sterling, Terry, McDonnell and Kim.)
- Published
- 2023
- Full Text
- View/download PDF
38. PHF1 is required for chromosome alignment and asymmetric division during mouse meiotic oocyte maturation.
- Author
-
Qu, Yi, Wang, Yang, and Qiao, Jie
- Abstract
In recent years, the etiological study of oocyte maturation failure and other mechanisms of early embryonic development has gradually advanced. However, while some achievements have been made in this field, the intrinsic mechanisms underlying disordered oocyte maturation remain unclear. Polycomb group proteins (PcG) are a family of proteins that are involved in the epigenetic silencing of genes. Many members of this family are reportedly involved in mammalian oocyte maturation and early embryonic development. PHD finger protein 1 (PHF1) is a core member of the polycomblike group of proteins, although its role in oocyte maturation and early embryonic development are unknown. A previous study by our group using single cell transcriptome analysis and high-throughput technology revealed that PHF1 mRNA was elevated in the human oocyte and the early preimplantation embryo. This suggests that PHF1 may play an important role in oocyte maturation and early embryonic development. In the present study, we aimed to reveal the biological function of PHF1 in mouse oocyte maturation and illuminate its regulatory mechanisms. We report here, for the first time, that PHF1 is necessary for the accurate alignment of chromosomes and oocyte euploidy, as well for the regulation of the asymmetric division of oocytes in mouse. The results of the present study may have the potential to provide a new research direction of human oocyte maturation disorder and early embryonic development block. These results may also provide new diagnosis or treatment strategies for clinical patients. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
39. DNA Repair and Mitotic Checkpoint Genes as Potential Predictors of Chemotherapy Response in Non-Small-Cell Lung Cancer
- Author
-
Rosell, Rafael, Taron, Miquel, Santarpia, Mariacarmela, Salazar, Fernanda, Ramirez, Jose Luis, Molina, Miguel Angel, Teicher, Beverly A., editor, and Innocenti, Federico, editor
- Published
- 2009
- Full Text
- View/download PDF
40. HDAC2/3 binding and deacetylation of BubR1 initiates spindle assembly checkpoint silencing.
- Author
-
Park, Inai, Kwon, Mi‐Sun, Paik, Sangjin, Kim, Hyeonjong, Lee, Hae‐Ock, Choi, Eunhee, and Lee, Hyunsook
- Subjects
- *
DEACETYLATION , *HISTONE deacetylase , *SPINDLE apparatus , *GENE silencing , *UBIQUITINATION , *BRCA genes - Abstract
BubR1 acetylation is essential in spindle assembly checkpoint ( SAC) signaling. Here we show that BubR1 deacetylation is a signal that initiates mitotic exit. Sustained BubR1 acetylation arrests the cells in metaphase, although chromosome congression is achieved. BubR1 deacetylation was coordinated with dephosphorylation in mitotic exit, suggesting the presence of a coordinated acetylation-phosphorylation code in mitotic signaling. Histone deacetylase ( HDAC) 2 and 3 bound to acetylated BubR1 exclusively in mitosis and led to the polyubiquitination of BubR1. Subsequent degradation of BubR1 resulted in the disassembly of the mitotic checkpoint complex. Importantly, BRCA2 was required for HDAC2/3 association with acetylated BubR1 in nocodazole (Noc)-arrested cells. Plk1, PP2A, P300/CBP-associated factor ( PCAF) and BubR1 were found in the mitotic BRCA2 complex, suggesting that BRCA2 acts as a signaling scaffold for BubR1 modification. Furthermore, we show that Plk1 is required for BRCA2 to localize at the prometaphase kinetochore (KT). Inhibition of Plk1 resulted in the loss of BRCA2 from the KT, and so did PCAF, consistent with the loss of BubR1 acetylation. Concordantly, BRCA2-dysfunctional cells exhibited resistance to trichostatin A, which was restored when BRCA2 was introduced. That loss of Brca2 conferred resistance to various HDAC inhibitors was corroborated by the experiments in mouse pancreatic organoids. These results suggest that the BRCA2-BubR1 acetylation-deacetylation pathway is an important decision-making point for the HDAC inhibitor response. Taken together, BRCA2 is a signaling platform for BubR1, and BubR1 deacetylation is a cue for SAC silencing. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
41. Cenp-meta is required for sustained spindle checkpoint
- Author
-
Thomas Rubin, Roger E. Karess, and Zohra Rahmani
- Subjects
Mitosis ,Spindle assembly checkpoint ,Metaphase ,Cenp-E ,BubR1 ,APC/C ,Kinetochore ,Science ,Biology (General) ,QH301-705.5 - Abstract
Cenp-E is a kinesin-like motor protein required for efficient end-on attachment of kinetochores to the spindle microtubules. Cenp-E immunodepletion in Xenopus mitotic extracts results in the loss of mitotic arrest and massive chromosome missegregation, whereas its depletion in mammalian cells leads to chromosome segregation defects despite the presence of a functional spindle assembly checkpoint (SAC). Cenp-meta has previously been reported to be the Drosophila homolog of vertebrate Cenp-E. In this study, we show that cenp-metaΔ mutant neuroblasts arrest in mitosis when treated with colchicine. cenp-metaΔ mutant cells display a mitotic delay. Yet, despite the persistence of the two checkpoint proteins Mad2 and BubR1 on unattached kinetochores, these cells eventually enter anaphase and give rise to highly aneuploid daughter cells. Indeed, we find that cenp-metaΔ mutant cells display a slow but continuous degradation of cyclin B, which eventually triggers the mitotic exit observed. Thus, our data provide evidence for a role of Cenp-meta in sustaining the SAC response.
- Published
- 2014
- Full Text
- View/download PDF
42. Is periapical surgery follow-up with only two-dimensional radiographs reliable? A retrospective cohort type sensitivity study
- Author
-
A. Ramis-Alario, B. Tarazona-Álvarez, M. Peñarrocha-Diago, D. Soto-Peñaloza, and D. Peñarrocha-Oltra
- Subjects
Cone beam computed tomography ,Radiography ,Computed tomography ,bubr1 ,oral leukoplakia ,Radiography, Panoramic ,epithelial dysplasia ,Humans ,Medicine ,General Dentistry ,Reference standards ,spindly ,UNESCO:CIENCIAS MÉDICAS ,Retrospective Studies ,medicine.diagnostic_test ,business.industry ,Research ,Periapical radiography ,Retrospective cohort study ,Cone-Beam Computed Tomography ,respiratory system ,bub3 ,mad2 ,oral squamous cell carcinoma ,Volume measurements ,Otorhinolaryngology ,Surgery ,Oral Surgery ,Nuclear medicine ,business ,Tooth ,Periapical surgery ,Follow-Up Studies - Abstract
Background Two-dimensional (2D) radiographic techniques are commonly used for assessing lesion prognosis after endodontic surgery. The present retrospective cohort study analyzes the sensitivity and ability of different radiographic techniques in obtaining area and volume measurements of periapical lesions. Material and Methods Preoperative and follow-up (6-48 months) periapical and panoramic radiographs (index test) and cone-beam computed tomography (CBCT) images (reference standard) were selected from an endodontic microsurgery database. Sensitivity was analyzed independently by two examiners. The areas of the 2D radiographic images and CBCT volumes were studied using Itk-Snap software and Romexis viewer. Results The sample comprised 105 patients and 105 teeth, with a mean follow-up of 21 months (range 6-48). Preoperatively, CBCT detected all the periapical areas, periapical radiography detected 67, and panoramic radiography detected 60. Postoperatively, of the 52 cases in which CBCT detected remains of the periapical area, periapical radiography detected 22, and panoramic radiography detected 17. The measurements of the areas obtained by the 2D methods, and the volumes obtained by CBCT, had to be transformed into linear measures for comparison purposes. The measurements were found to be significantly different in both the preoperative and the follow-up images. Conclusions Periapical radiography showed greater sensitivity than panoramic radiography, both preoperatively and at follow-up. The lesions measured with CBCT were larger, with significant differences than as evidenced by the periapical and panoramic radiographs. Key words:Periapical radiography, panoramic radiography, CBCT, sensitivity, treatment outcome, size of periapical radiolucency, area, volume.
- Published
- 2021
43. PP2A-B56γ is required for an efficient spindle assembly checkpoint.
- Author
-
Varadkar, Prajakta, Abbasi, Fatima, Takeda, Kazuyo, Dyson, Jade J., and McCright, Brent
- Abstract
The Spindle Assembly Checkpoint (SAC) is part of a complex feedback system designed to ensure that cells do not proceed through mitosis unless all chromosomal kinetochores have attached to spindle microtubules. The formation of the kinetochore complex and the implementation of the SAC are regulated by multiple kinases and phosphatases. BubR1 is a phosphoprotein that is part of the Cdc20 containing mitotic checkpoint complex that inhibits the APC/C so that Cyclin B1 and Securin are not degraded, thus preventing cells going into anaphase. In this study, we found that PP2A in association with its B56γ regulatory subunit, are needed for the stability of BubR1 during nocodazole induced cell cycle arrest. In primary cells that lack B56γ, BubR1 is prematurely degraded and the cells proceed through mitosis. The reduced SAC efficiency results in cells with abnormal chromosomal segregation, a hallmark of transformed cells. Previous studies on PP2A's role in the SAC and kinetochore formation were done using siRNAs to all 5 of the B56 family members. In our study we show that inactivation of only the PP2A-B56γ subunit can affect the efficiency of the SAC. We also provide data that show the intracellular locations of the B56 subunits varies between family members, which is consistent with the hypothesis that they are not completely functionally redundant. [ABSTRACT FROM PUBLISHER]
- Published
- 2017
- Full Text
- View/download PDF
44. Age-related decline in BubR1 impairs adult hippocampal neurogenesis.
- Author
-
Yang, Zhongxi, Jun, Heechul, Choi, Chan‐II, Yoo, Ki Hyun, Cho, Chang Hoon, Hussaini, Syed Mohammed Qasim, Simmons, Ambrosia J., Kim, Seonhee, Deursen, Jan M., Baker, Darren J., and Jang, Mi‐Hyeon
- Subjects
- *
DEVELOPMENTAL neurobiology , *AGING , *COGNITION disorders , *DENDRITES , *MORPHOGENESIS , *HIPPOCAMPUS (Brain) - Abstract
Aging causes significant declines in adult hippocampal neurogenesis and leads to cognitive disability. Emerging evidence demonstrates that decline in the mitotic checkpoint kinase BubR1 level occurs with natural aging and induces progeroid features in both mice and children with mosaic variegated aneuploidy syndrome. Whether BubR1 contributes to age-related deficits in hippocampal neurogenesis is yet to be determined. Here we report that BubR1 expression is significantly reduced with natural aging in the mouse brain. Using established progeroid mice expressing low amounts of BubR1, we demonstrate these mice exhibit deficits in neural progenitor proliferation and maturation, leading to reduction in new neuron production. Collectively, our identification of BubR1 as a new and critical factor controlling sequential steps across neurogenesis raises the possibility that BubR1 may be a key mediator regulating aging-related hippocampal pathology. Targeting BubR1 may represent a novel therapeutic strategy for age-related cognitive deficits. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
45. Different Functionality of Cdc20 Binding Sites within the Mitotic Checkpoint Complex.
- Author
-
Sewart, Katharina and Hauf, Silke
- Subjects
- *
CELL division , *BINDING sites , *GENETIC mutation , *EUKARYOTIC cells , *SEQUESTRATION (Chemistry) , *PHYSIOLOGY ,YEAST physiology - Abstract
Summary The mitotic checkpoint is a cellular safeguard that prevents chromosome missegregation in eukaryotic cells [ 1, 2 ]. Suboptimal functioning may foster chromosome missegregation in cancer cells [ 3 ]. Checkpoint signaling produces the “mitotic checkpoint complex” (MCC), which prevents anaphase by targeting Cdc20, the activator of the anaphase-promoting complex/cyclosome (APC/C). Recent biochemical and structural studies revealed that the human MCC binds two Cdc20 molecules, one (Cdc20 M ) through well-characterized, cooperative binding to Mad2 and Mad3/BubR1 (forming the “core MCC”) and the other one (Cdc20 A ) through additional binding sequences in Mad3/BubR1 [ 4–6 ]. Here, we dissect the different functionality of these sites in vivo. We show in fission yeast that, at low Cdc20 concentrations, Cdc20 M binding is sufficient for checkpoint activity and Cdc20 A binding becomes dispensable. Cdc20 A binding is mediated by the conserved Mad3 ABBA-KEN2-ABBA motif [ 7, 8 ], which we find additionally required for binding of the MCC to the APC/C and for MCC disassembly. Strikingly, deletion of the APC/C subunit Apc15 mimics mutations in this motif, revealing a shared function. This function of Apc15 may be masked in human cells by independent mediators of MCC-APC/C binding. Our data provide important in vivo support for the recent structure-based models and functionally dissect three elements of Cdc20 inhibition: (1) sequestration of Cdc20 in the core MCC, sufficient at low Cdc20 concentrations; (2) inhibition of a second Cdc20 through the Mad3 C terminus, independent of Mad2 binding to this Cdc20 molecule; and (3) occupancy of the APC/C with full MCC, where Mad3 and Apc15 are involved. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
46. The Mitotic Checkpoint Complex Requires an Evolutionary Conserved Cassette to Bind and Inhibit Active APC/C.
- Author
-
Di Fiore, Barbara, Wurzenberger, Claudia, Davey, Norman E., and Pines, Jonathon
- Subjects
- *
MITOSIS , *CHROMATIDS , *SPINDLE apparatus , *ANAPHASE , *CARRIER proteins , *N-terminal residues - Abstract
Summary The Spindle Assembly Checkpoint (SAC) ensures genomic stability by preventing sister chromatid separation until all chromosomes are attached to the spindle. It catalyzes the production of the Mitotic Checkpoint Complex (MCC), which inhibits Cdc20 to inactivate the Anaphase Promoting Complex/Cyclosome (APC/C). Here we show that two Cdc20-binding motifs in BubR1 of the recently identified ABBA motif class are crucial for the MCC to recognize active APC/C-Cdc20. Mutating these motifs eliminates MCC binding to the APC/C, thereby abolishing the SAC and preventing cells from arresting in response to microtubule poisons. These ABBA motifs flank a KEN box to form a cassette that is highly conserved through evolution, both in the arrangement and spacing of the ABBA-KEN-ABBA motifs, and association with the amino-terminal KEN box required to form the MCC. We propose that the ABBA-KEN-ABBA cassette holds the MCC onto the APC/C by binding the two Cdc20 molecules in the MCC-APC/C complex. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
47. Expanding the PP2A Interactome by Defining a B56-Specific SLiM.
- Author
-
Wang, Xinru, Bajaj, Rakhi, Bollen, Mathieu, Peti, Wolfgang, and Page, Rebecca
- Subjects
- *
CELLULAR signal transduction , *THREONINE , *ENZYME activation , *BINDING sites , *AMINO acid sequence - Abstract
Summary Specific interactions between proteins govern essential physiological processes including signaling. Many enzymes, especially the family of serine/threonine phosphatases (PSPs: PP1, PP2A, and PP2B/calcineurin/CN), recruit substrates and regulatory proteins by binding short linear motifs (SLiMs), short sequences found within intrinsically disordered regions that mediate specific protein-protein interactions. While tremendous progress had been made in identifying where and how SLiMs bind PSPs, especially PP1 and CN, essentially nothing is known about how SLiMs bind PP2A, a validated cancer drug target. Here we describe three structures of a PP2A-SLiM interaction (B56: pS -RepoMan, B56: pS -BubR1, and B56: pSpS -BubR1), show that this PP2A-specific SLiM is defined as LSPIxE, and then use these data to discover scores of likely PP2A regulators and substrates. Together, these data provide a powerful approach not only for dissecting PP2A interaction networks in cells but also for targeting PP2A diseases, such as cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
48. BUBR1 recruits PP2A via the B56 family of targeting subunits to promote chromosome congression
- Author
-
Peng Xu, Elizabeth A. Raetz, Mayumi Kitagawa, David M. Virshup, and Sang Hyun Lee
- Subjects
Aurora B ,B56-PP2A ,BUBR1 ,Chromosome congression ,Science ,Biology (General) ,QH301-705.5 - Abstract
Summary BUBR1 is a mitotic phosphoprotein essential for the maintenance of chromosome stability by promoting chromosome congression and proper kinetochore–microtubule (K-fiber) attachment, but the underlying mechanism(s) has remained elusive. Here we identify BUBR1 as a binding partner of the B56 family of Protein Phosphatase 2A regulatory subunits. The interaction between BUBR1 and the B56 family is required for chromosome congression, since point mutations in BUBR1 that block B56 binding abolish chromosome congression. The BUBR1:B56-PP2A complex opposes Aurora B kinase activity, since loss of the complex can be reverted by inhibiting Aurora B. Importantly, we show that the failure of BUBR1 to recruit B56-PP2A also contributes to the chromosome congression defects found in cells derived from patients with the Mosaic Variegated Aneuploidy (MVA) syndrome. Together, we propose that B56-PP2A is a key mediator of BUBR1's role in chromosome congression and functions by antagonizing Aurora B activity at the kinetochore for establishing stable kinetochore–microtubule attachment at the metaphase plate.
- Published
- 2013
- Full Text
- View/download PDF
49. BubR1 alterations that reinforce mitotic surveillance act against aneuploidy and cancer
- Author
-
Robbyn L Weaver, Jazeel F Limzerwala, Ryan M Naylor, Karthik B Jeganathan, Darren J Baker, and Jan M van Deursen
- Subjects
BubR1 ,aneuploidy ,cancer ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
BubR1 is a key component of the spindle assembly checkpoint (SAC). Mutations that reduce BubR1 abundance cause aneuploidization and tumorigenesis in humans and mice, whereas BubR1 overexpression protects against these. However, how supranormal BubR1 expression exerts these beneficial physiological impacts is poorly understood. Here, we used Bub1b mutant transgenic mice to explore the role of the amino-terminal (BubR1N) and internal (BubR1I) Cdc20-binding domains of BubR1 in preventing aneuploidy and safeguarding against cancer. BubR1N was necessary, but not sufficient to protect against aneuploidy and cancer. In contrast, BubR1 lacking the internal Cdc20-binding domain provided protection against both, which coincided with improved microtubule-kinetochore attachment error correction and SAC activity. Maximal SAC reinforcement occurred when both the Phe- and D-box of BubR1I were disrupted. Thus, while under- or overexpression of most mitotic regulators impairs chromosome segregation fidelity, certain manipulations of BubR1 can positively impact this process and therefore be therapeutically exploited.
- Published
- 2016
- Full Text
- View/download PDF
50. Suppression of Genomic Instabilities Caused by Chromosome Mis-segregation: A Perspective From Studying BubR1 and Sgo1
- Author
-
Wei Dai
- Subjects
BubR1 ,chromosome mis-segregation ,genomic instabilities ,Sgo1 ,Medicine (General) ,R5-920 - Abstract
Aneuploidy is a major manifestation of chromosomal instability, which is defined as a numerical abnormality of chromosomes in diploid cells. It is highly prevalent in a variety of human malignancies. Increased chromosomal instability is the major driving force for tumor development and progression. To suppress genomic stability during cell division, eukaryotic cells have evolved important molecular mechanisms, commonly referred to as checkpoints. The spindle checkpoint ensures that cells with defective mitotic spindles or a defective interaction between the spindles and kinetochores do not initiate chromosomal segregation during mitosis. Extensive studies have identified and characterized more than a dozen genes that play important roles in the regulation of the spindle checkpoint in mammalian cells. During the past decade, we have carried out extensive investigation of the role of BubR1 (Bub1-related kinase) and Sgo1 (shugoshin 1), two important gene products that safeguard accurate chromosome segregation during mitosis. This mini-review summarizes our studies, as well as those by other researchers in the field, on the functions of these two checkpoint proteins and their molecular regulation during mitosis. Further elucidation of the molecular mechanisms of the spindle checkpoint regulation has the potential to identify important mitotic targets for rational anticancer drug design.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.