1
|
Valles SY, Bural S, Godek KM, Compton DA. Cyclin A/Cdk1 promotes chromosome alignment and timely mitotic progression. Mol Biol Cell 2024; 35:ar141. [PMID: 39356777 PMCID: PMC11617097 DOI: 10.1091/mbc.e23-12-0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
To ensure genomic fidelity, a series of spatially and temporally coordinated events is executed during prometaphase of mitosis, including bipolar spindle formation, chromosome attachment to spindle microtubules at kinetochores, the correction of erroneous kinetochore-microtubule (k-MT) attachments, and chromosome congression to the spindle equator. Cyclin A/Cdk1 kinase plays a key role in destabilizing k-MT attachments during prometaphase to promote correction of erroneous k-MT attachments. However, it is unknown whether Cyclin A/Cdk1 kinase regulates other events during prometaphase. Here, we investigate additional roles of Cyclin A/Cdk1 in prometaphase by using an siRNA knockdown strategy to deplete endogenous Cyclin A from human cells. We find that depleting Cyclin A significantly extends mitotic duration, specifically prometaphase, because chromosome alignment is delayed. Unaligned chromosomes display erroneous monotelic, syntelic, or lateral k-MT attachments suggesting that bioriented k-MT attachment formation is delayed in the absence of Cyclin A. Mechanistically, chromosome alignment is likely impaired because the localization of the kinetochore proteins BUB1 kinase, KNL1, and MPS1 kinase are reduced in Cyclin A-depleted cells. Moreover, we find that Cyclin A promotes BUB1 kinetochore localization independently of its role in destabilizing k-MT attachments. Thus, Cyclin A/Cdk1 facilitates chromosome alignment during prometaphase to support timely mitotic progression.
Collapse
Affiliation(s)
- Sarah Y. Valles
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Shrea Bural
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Kristina M. Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Duane A. Compton
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| |
Collapse
|
2
|
Chen Q, Ouyang L, Liu Q. Cyclin B1: A potential prognostic and immunological biomarker in pan-cancer. BIOMOLECULES & BIOMEDICINE 2024; 24:1150-1169. [PMID: 38581717 PMCID: PMC11378994 DOI: 10.17305/bb.2024.10253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/08/2024]
Abstract
Cyclin B1 (CCNB1) encodes a regulatory protein essential for the regulation of cell mitosis, particularly in controlling the G2/M transition phase of the cell cycle. Current research has implicated CCNB1 in the progression of various types of cancer, including gastric cancer, breast cancer, and non-small cell lung cancer. In this study, we conducted a pan-cancer analysis of CCNB1 to investigate its prognostic significance and immunological aspects. Our comprehensive investigation covered a wide range of analyses, including gene expression, promoter methylation, genetic alterations, immune infiltration, immune regulators, and enrichment studies. We utilized multiple databases and tools for this purpose, such as The Cancer Genome Atlas (TCGA), the Genotype-Tissue Expression (GTEx) project, the Human Protein Atlas (HPA), the University of Alabama at Birmingham CANcer data analysis Portal (UALCAN), the Gene Expression Profiling Interactive Analysis (GEPIA), the DNA Methylation Interactive Visualization Database (DNMIVD), the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), Sangerbox, and cBioPortal. Data analyses were executed using GraphPad Prism software, R software, and various online tools. Our findings demonstrated a significant increase in CCNB1 expression across 28 cancer types. Elevated CCNB1 expression correlated with decreased overall survival (OS) in 11 cancer types and disease-free survival (DFS) in 12 cancer types. Additionally, DNA promoter methylation levels were significantly decreased in 14 cancer types. Furthermore, the study verified a significant association between CCNB1 expression and immune infiltration, immune modulators, microsatellite instability (MSI), and tumor mutational burden (TMB). Enrichment analysis indicated that CCNB1 is involved in multiple cellular pathways. Collectively, our results suggested that CCNB1 has the potential to serve as a valuable prognostic biomarker and may be a promising target for immunotherapy in various cancer types.
Collapse
Affiliation(s)
- Quan Chen
- Department of Pathology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hospital Department, Hubei University of Chinese Medicine, Wuhan, China
| | - Li Ouyang
- Department of Pathology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hospital Department, Hubei University of Chinese Medicine, Wuhan, China
| | - Qing Liu
- Department of Pathology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hospital Department, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
3
|
Shapiro JG, Changela N, Jang JK, Joshi JN, McKim KS. Distinct checkpoint and homolog biorientation pathways regulate meiosis I in Drosophila oocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608908. [PMID: 39229242 PMCID: PMC11370425 DOI: 10.1101/2024.08.21.608908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Mitosis and meiosis have two mechanisms for regulating the accuracy of chromosome segregation: error correction and the spindle assembly checkpoint (SAC). We have investigated the function of several checkpoint proteins in meiosis I of Drosophila oocytes. Evidence of a SAC response by several of these proteins is found upon depolymerization of microtubules by colchicine. However, unattached kinetochores or errors in biorientation of homologous chromosomes does not induce a SAC response. Furthermore, the metaphase I arrest does not depend on SAC genes, suggesting the APC is inhibited even if the SAC is silenced. Two SAC proteins, ROD of the ROD-ZW10-Zwilch (RZZ) complex and MPS1, are also required for the biorientation of homologous chromosomes during meiosis I, suggesting an error correction function. Both proteins aid in preventing or correcting erroneous attachments and depend on SPC105R for localization to the kinetochore. We have defined a region of SPC105R, amino acids 123-473, that is required for ROD localization and biorientation of homologous chromosomes at meiosis I. Surprisingly, ROD removal, or "streaming", is independent of the dynein adaptor Spindly and is not linked to the stabilization of end-on attachments. Instead, meiotic RZZ streaming appears to depend on cell cycle stage and may be regulated independently of kinetochore attachment or biorientation status. We also show that dynein adaptor Spindly is also required for biorientation at meiosis I, and surprisingly, the direction of RZZ streaming.
Collapse
Affiliation(s)
- Joanatta G Shapiro
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Neha Changela
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Janet K Jang
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Jay N Joshi
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Kim S McKim
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| |
Collapse
|
4
|
Huang H, Kung FL, Huang YW, Hsu CC, Guh JH, Hsu LC. Sensitization of cancer cells to paclitaxel-induced apoptosis by canagliflozin. Biochem Pharmacol 2024; 223:116140. [PMID: 38513740 DOI: 10.1016/j.bcp.2024.116140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
Cancer cells consume more glucose and usually overexpress glucose transporters which have become potential targets for the development of anticancer drugs. It has been demonstrated that selective SGLT2 inhibitors, such as canagliflozin and dapagliflozin, display anticancer activity. Here we demonstrated that canagliflozin and dapagliflozin synergistically enhanced the growth inhibitory effect of paclitaxel in cancer cells including ovarian cancer and oral squamous cell carcinoma cells. Canagliflozin also inhibited glucose uptake via GLUTs. The combination of paclitaxel and WZB117, a GLUT inhibitor, exhibited a strong synergy, supporting the notion that inhibition of GLUTs by canagliflozin may also account for the synergy between canagliflozin and paclitaxel. Mechanistic studies in ES-2 ovarian cancer cells revealed that canagliflozin potentiated paclitaxel-induced apoptosis and DNA damaging effect. Paclitaxel in the nanomolar range elevated abnormal mitotic cells as well as aneuploid cells, and canagliflozin further enhanced this effect. Furthermore, canagliflozin downregulated cyclin B1 and phospho-BUBR1 upon spindle assembly checkpoint (SAC) activation by paclitaxel, and may consequently impair SAC. Thus, paclitaxel disturbed microtubule dynamics and canagliflozin compromised SAC activity, together they may induce premature mitotic exit, accumulation of aneuploid cells with DNA damage, and ultimately apoptosis.
Collapse
Affiliation(s)
- Haoning Huang
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Fan-Lu Kung
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Yu-Wen Huang
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Chun-Chien Hsu
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Road, Taipei 10050, Taiwan.
| |
Collapse
|
5
|
Valles SY, Godek KM, Compton DA. Cyclin A/Cdk1 promotes chromosome alignment and timely mitotic progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572788. [PMID: 38187612 PMCID: PMC10769330 DOI: 10.1101/2023.12.21.572788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
To ensure genomic fidelity a series of spatially and temporally coordinated events are executed during prometaphase of mitosis, including bipolar spindle formation, chromosome attachment to spindle microtubules at kinetochores, the correction of erroneous kinetochore-microtubule (k-MT) attachments, and chromosome congression to the spindle equator. Cyclin A/Cdk1 kinase plays a key role in destabilizing k-MT attachments during prometaphase to promote correction of erroneous k-MT attachments. However, it is unknown if Cyclin A/Cdk1 kinase regulates other events during prometaphase. Here, we investigate additional roles of Cyclin A/Cdk1 in prometaphase by using an siRNA knockdown strategy to deplete endogenous Cyclin A from human cells. We find that depleting Cyclin A significantly extends mitotic duration, specifically prometaphase, because chromosome alignment is delayed. Unaligned chromosomes display erroneous monotelic, syntelic, or lateral k-MT attachments suggesting that bioriented k-MT attachment formation is delayed in the absence of Cyclin A. Mechanistically, chromosome alignment is likely impaired because the localization of the kinetochore proteins BUB1 kinase, KNL1, and MPS1 kinase are reduced in Cyclin A-depleted cells. Moreover, we find that Cyclin A promotes BUB1 kinetochore localization independently of its role in destabilizing k-MT attachments. Thus, Cyclin A/Cdk1 facilitates chromosome alignment during prometaphase to support timely mitotic progression.
Collapse
Affiliation(s)
- Sarah Y Valles
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kristina M Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Duane A Compton
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
6
|
Zhu J, Li X, Lv F, Zhou W. Bioinformatics Approach to Identify the Influences of COVID-19 on Ischemic Stroke. Biochem Genet 2023; 61:2222-2241. [PMID: 37184686 PMCID: PMC10184096 DOI: 10.1007/s10528-023-10366-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/09/2023] [Indexed: 05/16/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) is becoming more infectious and less virulent, symptoms beyond the lungs of the Coronavirus Disease 2019 (COVID-19) patients are a growing concern. Studies have found that the severity of COVID-19 patients is associated with an increased risk of ischemic stroke (IS); however, the underlying pathogenic mechanisms remain unknown. In this study, bioinformatics approaches were utilized to explore potential pathogenic mechanisms and predict potential drugs that may be useful in the treatment of COVID-19 and IS. The GSE152418 and GSE122709 datasets were downloaded from the GEO website to obtain the common differentially expressed genes (DEGs) of the two datasets for further functional enrichment, pathway analysis, and drug candidate prediction. A total of 80 common DEGs were identified in COVID-19 and IS datasets for GO and KEGG analysis. Next, the protein-protein interaction (PPI) network was constructed and hub genes were identified. Further, transcription factor-gene interactions and DEGs-miRNAs coregulatory network were investigated to explore their regulatory roles in disease. Finally, protein-drug interactions with common DEGs were analyzed to predict potential drugs. We successfully identified the top 10 hub genes that could serve as novel targeted therapies for COVID-19 and screened out some potential drugs for the treatment of COVID-19 and IS.
Collapse
Affiliation(s)
- Jiabao Zhu
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China
| | - Xiangui Li
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China
| | - Fanzhen Lv
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China
| | - Weimin Zhou
- Department of Vascular Surgery, The Second Affliated Hospital of Nanchang University, Minde Road 1, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
7
|
Qi X, Liu Y, Peng Y, Fu Y, Fu Y, Yin L, Li X. UHRF1 promotes spindle assembly and chromosome congression by catalyzing EG5 polyubiquitination. J Cell Biol 2023; 222:e202210093. [PMID: 37728657 PMCID: PMC10510743 DOI: 10.1083/jcb.202210093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 05/18/2023] [Accepted: 08/24/2023] [Indexed: 09/21/2023] Open
Abstract
UHRF1 is an epigenetic coordinator bridging DNA methylation and histone modifications. Additionally, UHRF1 regulates DNA replication and cell cycle, and its deletion induces G1/S or G2/M cell cycle arrest. The roles of UHRF1 in the regulation of G2/M transition remain poorly understood. UHRF1 depletion caused chromosome misalignment, thereby inducing cell cycle arrest at mitotic metaphase, and these cells exhibited the defects of spindle geometry, prominently manifested as shorter spindles. Mechanistically, UHRF1 protein directly interacts with EG5, a kinesin motor protein, during mitosis. Furthermore, UHRF1 induced EG5 polyubiquitination at the site of K1034 and further promoted the interaction of EG5 with spindle assembly factor TPX2, thereby ensuring accurate EG5 distribution to the spindles during metaphase. Our study clarifies a novel UHRF1 function as a nuclear protein catalyzing EG5 polyubiquitination for proper spindle architecture and faithful genomic transmission, which is independent of its roles in epigenetic regulation and DNA damage repair inside the nucleus. These findings revealed a previously unknown mechanism of UHRF1 in controlling mitotic spindle architecture and chromosome behavior and provided mechanistic evidence for UHRF1 deletion-mediated G2/M arrest.
Collapse
Affiliation(s)
- Xuli Qi
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youhong Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuchong Peng
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuxin Fu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongming Fu
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Linglong Yin
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiong Li
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
8
|
MacKenzie A, Vicory V, Lacefield S. Meiotic cells escape prolonged spindle checkpoint activity through kinetochore silencing and slippage. PLoS Genet 2023; 19:e1010707. [PMID: 37018287 PMCID: PMC10109492 DOI: 10.1371/journal.pgen.1010707] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/17/2023] [Accepted: 03/20/2023] [Indexed: 04/06/2023] Open
Abstract
To prevent chromosome mis-segregation, a surveillance mechanism known as the spindle checkpoint delays the cell cycle if kinetochores are not attached to spindle microtubules, allowing the cell additional time to correct improper attachments. During spindle checkpoint activation, checkpoint proteins bind the unattached kinetochore and send a diffusible signal to inhibit the anaphase promoting complex/cyclosome (APC/C). Previous work has shown that mitotic cells with depolymerized microtubules can escape prolonged spindle checkpoint activation in a process called mitotic slippage. During slippage, spindle checkpoint proteins bind unattached kinetochores, but the cells cannot maintain the checkpoint arrest. We asked if meiotic cells had as robust of a spindle checkpoint response as mitotic cells and whether they also undergo slippage after prolonged spindle checkpoint activity. We performed a direct comparison between mitotic and meiotic budding yeast cells that signal the spindle checkpoint through two different assays. We find that the spindle checkpoint delay is shorter in meiosis I or meiosis II compared to mitosis, overcoming a checkpoint arrest approximately 150 minutes earlier in meiosis than in mitosis. In addition, cells in meiosis I escape spindle checkpoint signaling using two mechanisms, silencing the checkpoint at the kinetochore and through slippage. We propose that meiotic cells undertake developmentally-regulated mechanisms to prevent persistent spindle checkpoint activity to ensure the production of gametes.
Collapse
Affiliation(s)
- Anne MacKenzie
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Victoria Vicory
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Soni Lacefield
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
- Department of Biochemistry and Cell Biology, the Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
9
|
Serpico AF, Pisauro C, Grieco D. On the assembly of the mitotic spindle, bistability and hysteresis. Cell Mol Life Sci 2023; 80:83. [PMID: 36890394 PMCID: PMC9995516 DOI: 10.1007/s00018-023-04727-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 03/10/2023]
Abstract
During cell division, the transition from interphase to mitosis is dictated by activation of the cyclin B-cdk1 (Cdk1) complex, master mitotic kinase. During interphase, Cdk1 accumulates in an inactive state (pre-Cdk1). When Cdk1 overcomes a certain threshold of activity upon initial activation of pre-Cdk1, then the stockpiled pre-Cdk1 is rapidly converted into overshooting active Cdk1, and mitosis is established irreversibly in a switch-like fashion. This is granted by positive Cdk1 activation loops and the concomitant inactivation of Cdk1 counteracting phosphatases, empowering Cdk1 activity and favoring the Cdk1-dependent phosphorylations that are required to establish mitosis. These circuitries prevent backtracking and ensure unidirectionality so that interphase and mitosis are considered bistable states. Mitosis also shows hysteresis, meaning that the levels of Cdk1 activity needed to establish mitosis are higher than those required to maintain it; therefore, once in mitosis cells can tolerate moderate drops in Cdk1 activity without exiting mitosis. Whether these features have other functional implications in addition to the general action of preventing backtracking is unknown. Here, we contextualize these concepts in the view of recent evidence indicating that loss of activity of small and compartmentalized amounts of Cdk1 within mitosis is necessary to assemble the mitotic spindle, the structure required to segregate replicated chromosomes. We further propose that, in addition to prevent backtracking, the stability and hysteresis properties of mitosis are also essential to move forward in mitosis by allowing cells to bear small, localized, drops in Cdk1 activity that are necessary to build the mitotic spindle.
Collapse
Affiliation(s)
| | | | - Domenico Grieco
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy. .,DMMBM, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
10
|
Rossi M, De Martino V, Di Giuseppe L, Battafarano G, Di Gregorio J, Terreri S, Marampon F, Minisola S, Del Fattore A. Anti-proliferative, pro-apototic and anti-migratory properties of HDAC inhibitor PXD-101 on osteosarcoma cell lines. Arch Biochem Biophys 2023; 734:109489. [PMID: 36526001 DOI: 10.1016/j.abb.2022.109489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
The therapeutic strategies for osteosarcoma involve both surgical approach and chemotherapy, but the identification of new therapeutic targets is particularly necessary in patients with local chemo-resistance, recurrence and lung metastases. The role of epigenetic regulation in osteosarcoma is largely unknown. Thus, in this study we disclosed the effects of histone deacetylase inhibitor drug PXD-101 on human osteosarcoma (OS) cell lines with different aggressiveness, including Saos-2, HOS and 143B cell lines. XTT assays revealed that treatment of Saos-2, HOS and 143B cells with PXD-101 decreased cell viability in a concentration-dependent manner. Fluorescence-activated cell sorting (FACS) analysis showed that PXD-101 inhibited proliferation and induced cell apoptosis. Wound healing assay indicated that PXD-101 inhibited migration of osteosarcoma cells. Real-Time RT-qPCR and protein analysis highlighted reduced expression of Runx2, Osterix and Mad2, probably due to Cyclin B1 inhibition by PXD-101 treatment. To our knowledge, this is the first study that characterized the anti-tumoral effect of PXD-101 in OS cells, suggesting a potential new therapeutic approach in osteosarcoma patients.
Collapse
Affiliation(s)
- Michela Rossi
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children's Hospital, IRCCS, viale San Paolo 15, 00146, Rome, Italy.
| | - Viviana De Martino
- Department of Clinical, Internal, Anaesthesiological and Cardiovascular Sciences, "Sapienza" University, viale del Policlinico 155, 00161, Rome, Italy.
| | - Laura Di Giuseppe
- Department of Clinical, Internal, Anaesthesiological and Cardiovascular Sciences, "Sapienza" University, viale del Policlinico 155, 00161, Rome, Italy.
| | - Giulia Battafarano
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children's Hospital, IRCCS, viale San Paolo 15, 00146, Rome, Italy.
| | - Jacopo Di Gregorio
- Department of Clinical, Internal, Anaesthesiological and Cardiovascular Sciences, "Sapienza" University, viale del Policlinico 155, 00161, Rome, Italy.
| | - Sara Terreri
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children's Hospital, IRCCS, viale San Paolo 15, 00146, Rome, Italy.
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University, viale del Policlinico 155, 00161, Rome, Italy.
| | - Salvatore Minisola
- Department of Clinical, Internal, Anaesthesiological and Cardiovascular Sciences, "Sapienza" University, viale del Policlinico 155, 00161, Rome, Italy.
| | - Andrea Del Fattore
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children's Hospital, IRCCS, viale San Paolo 15, 00146, Rome, Italy.
| |
Collapse
|
11
|
Hayward D, Roberts E, Gruneberg U. MPS1 localizes to end-on microtubule-attached kinetochores to promote microtubule release. Curr Biol 2022; 32:5200-5208.e8. [PMID: 36395767 DOI: 10.1016/j.cub.2022.10.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/07/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022]
Abstract
In eukaryotes, the spindle assembly checkpoint protects genome stability in mitosis by preventing chromosome segregation until incorrect microtubule-kinetochore attachment geometries have been eliminated and chromosome biorientation has been completed. These error correction and checkpoint processes are linked by the conserved Aurora B and MPS1 Ser/Thr kinases.1,2 MPS1-dependent checkpoint signaling is believed to be initiated by kinetochores without end-on microtubule attachments,3,4 including those generated by Aurora B-mediated error correction. The current model posits that MPS1 competes with microtubules for binding sites at the kinetochore.3,4 MPS1 is thought to first recognize kinetochores not blocked by microtubules and then initiate checkpoint signaling. However, MPS1 is also required for chromosome biorientation and correction of microtubule-kinetochore attachment errors.5,6,7,8,9 This latter function, which must require direct interaction with microtubule-attached kinetochores, is not readily explained within the constraints of the current model. Here, we show that MPS1 transiently localizes to end-on attached kinetochores and that this recruitment depends on the relative activities of Aurora B and its counteracting phosphatase PP2A-B56 rather than microtubule-attachment state per se. MPS1 autophosphorylation also regulates MPS1 kinetochore levels but does not determine the response to microtubule attachment. At end-on attached kinetochores, MPS1 actively promotes microtubule release together with Aurora B. Furthermore, in live cells, MPS1 is detected at attached kinetochores before the removal of microtubules. During chromosome alignment, MPS1, therefore, coordinates both the resolution of incorrect microtubule-kinetochore attachments and the initiation of spindle checkpoint signaling.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3RE, UK
| | - Emile Roberts
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3RE, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3RE, UK.
| |
Collapse
|
12
|
Cao X, Shami Shah A, Sanford EJ, Smolka MB, Baskin JM. Proximity Labeling Reveals Spatial Regulation of the Anaphase-Promoting Complex/Cyclosome by a Microtubule Adaptor. ACS Chem Biol 2022; 17:2605-2618. [PMID: 35952650 PMCID: PMC9933862 DOI: 10.1021/acschembio.2c00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The anaphase-promoting complex/cyclosome (APC/C) coordinates advancement through mitosis via temporally controlled polyubiquitination events. Despite the long-appreciated spatial organization of key events in mitosis mediated largely by cytoskeletal networks, the spatial regulation of APC/C, the major mitotic E3 ligase, is poorly understood. We describe a microtubule-resident protein, PLEKHA5, as an interactor of APC/C and spatial regulator of its activity in mitosis. Microtubule-localized proximity biotinylation tools revealed that PLEKHA5 depletion decreased APC/C association with the microtubule cytoskeleton, which prevented efficient loading of APC/C with its coactivator CDC20 and led to reduced APC/C E3 ligase activity. PLEKHA5 knockdown delayed mitotic progression, causing accumulation of APC/C substrates dependent upon the PLEKHA5-APC/C interaction in microtubules. We propose that PLEKHA5 functions as an adaptor of APC/C that promotes its subcellular localization to microtubules and facilitates its activation by CDC20, thus ensuring the timely turnover of key mitotic APC/C substrates and proper progression through mitosis.
Collapse
Affiliation(s)
- Xiaofu Cao
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, United States
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14850, United States
| | - Adnan Shami Shah
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, United States
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14850, United States
| | - Ethan J Sanford
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14850, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14850, United States
| | - Marcus B Smolka
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14850, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14850, United States
| | - Jeremy M Baskin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, United States
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14850, United States
| |
Collapse
|
13
|
Liu S, Yuan X, Gui P, Liu R, Durojaye O, Hill DL, Fu C, Yao X, Dou Z, Liu X. Mad2 promotes Cyclin B2 recruitment to the kinetochore for guiding accurate mitotic checkpoint. EMBO Rep 2022; 23:e54171. [PMID: 35384228 PMCID: PMC9171689 DOI: 10.15252/embr.202154171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
Accurate mitotic progression relies on the dynamic phosphorylation of multiple substrates by key mitotic kinases. Cyclin-dependent kinase 1 is a master kinase that coordinates mitotic progression and requires its regulatory subunit Cyclin B to ensure full kinase activity and substrate specificity. The function of Cyclin B2, which is a closely related family member of Cyclin B1, remains largely elusive. Here, we show that Mad2 promotes the kinetochore localization of Cyclin B2 and that their interaction at the kinetochores guides accurate chromosome segregation. Our biochemical analyses have characterized the Mad2-Cyclin B2 interaction and delineated a novel Mad2-interacting motif (MIM) on Cyclin B2. The functional importance of the Cyclin B2-Mad2 interaction was demonstrated by real-time imaging in which MIM-deficient mutant Cyclin B2 failed to rescue the chromosomal segregation defects. Taken together, we have delineated a previously undefined function of Cyclin B2 at the kinetochore and have established, in human cells, a mechanism of action by which Mad2 contributes to the spindle checkpoint.
Collapse
Affiliation(s)
- Sikai Liu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Yuan
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ping Gui
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ran Liu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Olanrewaju Durojaye
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Donald L Hill
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zhen Dou
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
14
|
Compartmentalized control of Cdk1 drives mitotic spindle assembly. Cell Rep 2022; 38:110305. [DOI: 10.1016/j.celrep.2022.110305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/14/2021] [Accepted: 01/06/2022] [Indexed: 12/28/2022] Open
|
15
|
Rong MH, Li JD, Zhong LY, Huang YZ, Chen J, Xie LY, Qin RX, He XL, Zhu ZH, Huang SN, Zhou XG. CCNB1 promotes the development of hepatocellular carcinoma by mediating DNA replication in the cell cycle. Exp Biol Med (Maywood) 2021; 247:395-408. [PMID: 34743578 DOI: 10.1177/15353702211049149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In our studies, cyclin B1 (CCNB1) mRNA and protein were overexpressed in hepatocellular carcinoma (HCC) tissues compared with non-HCC tissues. Moreover, CCNB1 was overexpressed in the serum of HCC patients. The expression of CCNB1 was associated with several crucial clinicopathologic characteristics, and the HCC patients with overexpressed CCNB1 had worse overall survival outcomes. In the screening of interactional genes, a total of 266 upregulated co-expression genes, which were positively associated with CCNB1, were selected from the datasets, and 67 downregulated co-expression genes, which were negatively associated with CCNB1, were identified. The key genes might be functionally enriched in DNA replication and the cell cycle pathways. CDC20, CCNA2, PLK1, and FTCD were selected for further research because they were highly connected in the protein-protein interaction networks. Upregulated CDC20, CCNA2, and PLK1 and downregulated FTCD might result in undesirable overall survival outcomes for HCC patients. The univariate Cox analysis results showed that CDC20 and PLK1 might be two independent risk factors, while FTCD might be protective in HCC. Therefore, CCNB1 may participate in the cell cycle of HCC by regulating DNA replication, and CCNB1 may provide a direction for the diagnosis of early-stage HCC and targeted HCC therapy.
Collapse
Affiliation(s)
- Min-Hua Rong
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jian-Di Li
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Lu-Yang Zhong
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yu-Zhen Huang
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Juan Chen
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Li-Yuan Xie
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rong-Xing Qin
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiao-Lian He
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhan-Hui Zhu
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Su-Ning Huang
- Department of Radiotherapy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xian-Guo Zhou
- Research Department, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
16
|
Lara-Gonzalez P, Pines J, Desai A. Spindle assembly checkpoint activation and silencing at kinetochores. Semin Cell Dev Biol 2021; 117:86-98. [PMID: 34210579 PMCID: PMC8406419 DOI: 10.1016/j.semcdb.2021.06.009] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 01/01/2023]
Abstract
The spindle assembly checkpoint (SAC) is a surveillance mechanism that promotes accurate chromosome segregation in mitosis. The checkpoint senses the attachment state of kinetochores, the proteinaceous structures that assemble onto chromosomes in mitosis in order to mediate their interaction with spindle microtubules. When unattached, kinetochores generate a diffusible inhibitor that blocks the activity of the anaphase-promoting complex/cyclosome (APC/C), an E3 ubiquitin ligase required for sister chromatid separation and exit from mitosis. Work from the past decade has greatly illuminated our understanding of the mechanisms by which the diffusible inhibitor is assembled and how it inhibits the APC/C. However, less is understood about how SAC proteins are recruited to kinetochores in the absence of microtubule attachment, how the kinetochore catalyzes formation of the diffusible inhibitor, and how attachments silence the SAC at the kinetochore. Here, we summarize current understanding of the mechanisms that activate and silence the SAC at kinetochores and highlight open questions for future investigation.
Collapse
Affiliation(s)
- Pablo Lara-Gonzalez
- Ludwig Institute for Cancer Research, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | | | - Arshad Desai
- Ludwig Institute for Cancer Research, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
17
|
Mengoli V, Jonak K, Lyzak O, Lamb M, Lister LM, Lodge C, Rojas J, Zagoriy I, Herbert M, Zachariae W. Deprotection of centromeric cohesin at meiosis II requires APC/C activity but not kinetochore tension. EMBO J 2021; 40:e106812. [PMID: 33644894 PMCID: PMC8013787 DOI: 10.15252/embj.2020106812] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 01/03/2023] Open
Abstract
Genome haploidization involves sequential loss of cohesin from chromosome arms and centromeres during two meiotic divisions. At centromeres, cohesin's Rec8 subunit is protected from separase cleavage at meiosis I and then deprotected to allow its cleavage at meiosis II. Protection of centromeric cohesin by shugoshin-PP2A seems evolutionarily conserved. However, deprotection has been proposed to rely on spindle forces separating the Rec8 protector from cohesin at metaphase II in mammalian oocytes and on APC/C-dependent destruction of the protector at anaphase II in yeast. Here, we have activated APC/C in the absence of sister kinetochore biorientation at meiosis II in yeast and mouse oocytes, and find that bipolar spindle forces are dispensable for sister centromere separation in both systems. Furthermore, we show that at least in yeast, protection of Rec8 by shugoshin and inhibition of separase by securin are both required for the stability of centromeric cohesin at metaphase II. Our data imply that related mechanisms preserve the integrity of dyad chromosomes during the short metaphase II of yeast and the prolonged metaphase II arrest of mammalian oocytes.
Collapse
Affiliation(s)
- Valentina Mengoli
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
Institute for Research in BiomedicineUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Katarzyna Jonak
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Oleksii Lyzak
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Mahdi Lamb
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Lisa M Lister
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Chris Lodge
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Julie Rojas
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Ievgeniia Zagoriy
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
EMBL HeidelbergHeidelbergGermany
| | - Mary Herbert
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Wolfgang Zachariae
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
18
|
Liu C, Dai Q, Ding Q, Wei M, Kong X. Identification of key genes in hepatitis B associated hepatocellular carcinoma based on WGCNA. Infect Agent Cancer 2021; 16:18. [PMID: 33726794 PMCID: PMC7962393 DOI: 10.1186/s13027-021-00357-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic Infection of Hepatitis B virus (HBV) is one risk factor of hepatocellular carcinoma (HCC). Much effort has been made to research the process of HBV-associated HCC, but its molecular mechanisms of carcinogenesis remain vague. Here, weighted gene co-expression network analysis (WGCNA) was employed to explore the co-expressed modules and hub/key genes correlated to HBV-associated HCC. We found that genes of the most significant module related to HBV-associated HCC were enriched in DNA replication, p53 signaling pathway, cell cycle, and HTLV-1 infection associated pathway; these cellular pathways played critical roles in the initiation and development of HCC or viral infections. Furthermore, seven hub/key genes were identified based on the topological network analysis, and their roles in HCC were verified by expression and Kaplan-Meier survival analysis. Protein-protein interaction and KEGG pathway analysis suggested that these key genes may stimulate cellular proliferation to promote the HCC progression. This study provides new perspectives to the knowledge of the key pathways and genes in the carcinogenesis process of HBV-associated HCC, and our findings provided potential therapeutic targets and clues of the carcinogenesis of HBV-associated HCC.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China.
| | - Qinghai Dai
- Nankai University Second People's Hospital, Nankai University, Tianjin, China
| | - Qian Ding
- School of Medicine, Nankai University, Tianjin, China
| | - Min Wei
- School of Medicine, Nankai University, Tianjin, China. .,Nankai University Second People's Hospital, Nankai University, Tianjin, China.
| | - Xiaohong Kong
- School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
19
|
The PHLPP1 N-Terminal Extension Is a Mitotic Cdk1 Substrate and Controls an Interactome Switch. Mol Cell Biol 2021; 41:e0033320. [PMID: 33397691 PMCID: PMC8088274 DOI: 10.1128/mcb.00333-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
PH domain leucine-rich repeat protein phosphatase 1 (PHLPP1) is a tumor suppressor that directly dephosphorylates a wide array of substrates, most notably the prosurvival kinase Akt. However, little is known about the molecular mechanisms governing PHLPP1 itself. Here, we report that PHLPP1 is dynamically regulated in a cell cycle-dependent manner and deletion of PHLPP1 results in mitotic delays and increased rates of chromosomal segregation errors. We show that PHLPP1 is hyperphosphorylated during mitosis by Cdk1 in a functionally uncharacterized region known as the PHLPP1 N-terminal extension (NTE). A proximity-dependent biotin identification (BioID) interaction screen revealed that during mitosis, PHLPP1 dissociates from plasma membrane scaffolds, such as Scribble, by a mechanism that depends on its NTE and gains proximity to kinetochore and mitotic spindle proteins such as KNL1 and TPX2. Our data are consistent with a model in which phosphorylation of PHLPP1 during mitosis regulates binding to its mitotic partners and allows accurate progression through mitosis. The finding that PHLPP1 binds mitotic proteins in a cell cycle- and phosphorylation-dependent manner may have relevance to its tumor-suppressive function.
Collapse
|
20
|
Specificity determinants of phosphoprotein phosphatases controlling kinetochore functions. Essays Biochem 2021; 64:325-336. [PMID: 32501472 DOI: 10.1042/ebc20190065] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022]
Abstract
Kinetochores are instrumental for accurate chromosome segregation by binding to microtubules in order to move chromosomes and by delaying anaphase onset through the spindle assembly checkpoint (SAC). Dynamic phosphorylation of kinetochore components is key to control these activities and is tightly regulated by temporal and spatial recruitment of kinases and phosphoprotein phosphatases (PPPs). Here we focus on PP1, PP2A-B56 and PP2A-B55, three PPPs that are important regulators of mitosis. Despite the fact that these PPPs share a very similar active site, they target unique ser/thr phosphorylation sites to control kinetochore function. Specificity is in part achieved by PPPs binding to short linear motifs (SLiMs) that guide their substrate specificity. SLiMs bind to conserved pockets on PPPs and are degenerate in nature, giving rise to a range of binding affinities. These SLiMs control the assembly of numerous substrate specifying complexes and their position and binding strength allow PPPs to target specific phosphorylation sites. In addition, the activity of PPPs is regulated by mitotic kinases and inhibitors, either directly at the activity level or through affecting PPP-SLiM interactions. Here, we discuss recent progress in understanding the regulation of PPP specificity and activity and how this controls kinetochore biology.
Collapse
|
21
|
Sarkar D, Diermeier SD. Circular RNAs: Potential Applications as Therapeutic Targets and Biomarkers in Breast Cancer. Noncoding RNA 2021; 7:2. [PMID: 33466455 PMCID: PMC7838774 DOI: 10.3390/ncrna7010002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/26/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
Circular RNAs (circRNAs) are a class of non-coding RNAs that form a covalently closed loop. A number of functions and mechanisms of action for circRNAs have been reported, including as miRNA sponge, exerting transcriptional and translational regulation, interacting with proteins, and coding for peptides. CircRNA dysregulation has also been implicated in many cancers, such as breast cancer. Their relatively high stability and presence in bodily fluids makes cancer-associated circRNAs promising candidates as a new biomarker. In this review, we summarize the research undertaken on circRNAs associated with breast cancer, discuss circRNAs as biomarkers, and present circRNA-based therapeutic approaches.
Collapse
Affiliation(s)
| | - Sarah D. Diermeier
- Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand;
| |
Collapse
|
22
|
BUBR1 Pseudokinase Domain Promotes Kinetochore PP2A-B56 Recruitment, Spindle Checkpoint Silencing, and Chromosome Alignment. Cell Rep 2020; 33:108397. [PMID: 33207204 DOI: 10.1016/j.celrep.2020.108397] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 10/13/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
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.
Collapse
|
23
|
Bancroft J, Holder J, Geraghty Z, Alfonso-Pérez T, Murphy D, Barr FA, Gruneberg U. PP1 promotes cyclin B destruction and the metaphase-anaphase transition by dephosphorylating CDC20. Mol Biol Cell 2020; 31:2315-2330. [PMID: 32755477 PMCID: PMC7851957 DOI: 10.1091/mbc.e20-04-0252] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/30/2022] Open
Abstract
Ubiquitin-dependent proteolysis of cyclin B and securin initiates sister chromatid segregation and anaphase. The anaphase-promoting complex/cyclosome and its coactivator CDC20 (APC/CCDC20) form the main ubiquitin E3 ligase for these two proteins. APC/CCDC20 is regulated by CDK1-cyclin B and counteracting PP1 and PP2A family phosphatases through modulation of both activating and inhibitory phosphorylation. Here, we report that PP1 promotes cyclin B destruction at the onset of anaphase by removing specific inhibitory phosphorylation in the N-terminus of CDC20. Depletion or chemical inhibition of PP1 stabilizes cyclin B and results in a pronounced delay at the metaphase-to-anaphase transition after chromosome alignment. This requirement for PP1 is lost in cells expressing CDK1 phosphorylation-defective CDC206A mutants. These CDC206A cells show a normal spindle checkpoint response and rapidly destroy cyclin B once all chromosomes have aligned and enter into anaphase in the absence of PP1 activity. PP1 therefore facilitates the metaphase-to-anaphase transition by promoting APC/CCDC20-dependent destruction of cyclin B in human cells.
Collapse
Affiliation(s)
- James Bancroft
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - James Holder
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Zoë Geraghty
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | - Daniel Murphy
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| |
Collapse
|
24
|
Cunha-Silva S, Conde C. From the Nuclear Pore to the Fibrous Corona: A MAD Journey to Preserve Genome Stability. Bioessays 2020; 42:e2000132. [PMID: 32885448 DOI: 10.1002/bies.202000132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/22/2020] [Indexed: 11/09/2022]
Abstract
The relationship between kinetochores and nuclear pore complexes (NPCs) is intimate but poorly understood. Several NPC components and associated proteins are relocated to mitotic kinetochores to assist in different activities that ensure faithful chromosome segregation. Such is the case of the Mad1-c-Mad2 complex, the catalytic core of the spindle assembly checkpoint (SAC), a surveillance pathway that delays anaphase until all kinetochores are attached to spindle microtubules. Mad1-c-Mad2 is recruited to discrete domains of unattached kinetochores from where it promotes the rate-limiting step in the assembly of anaphase-inhibitory complexes. SAC proficiency further requires Mad1-c-Mad2 to be anchored at NPCs during interphase. However, the mechanistic relevance of this arrangement for SAC function remains ill-defined. Recent studies uncover the molecular underpinnings that coordinate the release of Mad1-c-Mad2 from NPCs with its prompt recruitment to kinetochores. Here, current knowledge on Mad1-c-Mad2 function and spatiotemporal regulation is reviewed and the critical questions that remain unanswered are highlighted.
Collapse
Affiliation(s)
- Sofia Cunha-Silva
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal.,Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, 4050-313, Portugal
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| |
Collapse
|
25
|
Kuijt TEF, Lambers MLA, Weterings S, Ponsioen B, Bolhaqueiro ACF, Staijen DHM, Kops GJPL. A Biosensor for the Mitotic Kinase MPS1 Reveals Spatiotemporal Activity Dynamics and Regulation. Curr Biol 2020; 30:3862-3870.e6. [PMID: 32888483 DOI: 10.1016/j.cub.2020.07.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/18/2020] [Accepted: 07/14/2020] [Indexed: 01/10/2023]
Abstract
Accurate chromosome segregation during cell division critically depends on error correction of chromosome-spindle interactions and the spindle assembly checkpoint (SAC) [1-3]. The kinase MPS1 is an essential regulator of both processes, ensuring full chromosome biorientation before anaphase onset [3, 4]. To understand when and where MPS1 activation occurs and how MPS1 signaling is modulated during mitosis, we developed MPS1sen, a sensitive and specific FRET-based biosensor for MPS1 activity. By placing MPS1sen at different subcellular locations, we show that MPS1 activity initiates in the nucleus ∼9-12 min prior to nuclear envelope breakdown (NEB) in a kinetochore-dependent manner and reaches the cytoplasm at the start of NEB. Soon after initiation, MPS1 activity increases with switch-like kinetics, peaking at completion of NEB. We further show that timing and extent of pre-NEB MPS1 activity is regulated by Aurora B and PP2A-B56. MPS1sen phosphorylation declines in prometaphase as a result of formation of kinetochore-microtubule attachments, reaching low but still detectable levels at metaphase. Finally, leveraging the sensitivity and dynamic range of MPS1sen, we show deregulated MPS1 signaling dynamics in colorectal cancer cell lines and tumor organoids with diverse genomic instability phenotypes.
Collapse
Affiliation(s)
- Timo E F Kuijt
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Maaike L A Lambers
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Sonja Weterings
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Bas Ponsioen
- Cancer Genomics Netherlands, UMC Utrecht, 3584CG Utrecht, the Netherlands; Molecular Cancer Research, Centre for Molecular Medicine, UMC Utrecht, 3584CG Utrecht, the Netherlands
| | - Ana C F Bolhaqueiro
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Debbie H M Staijen
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands.
| |
Collapse
|
26
|
Holder J, Mohammed S, Barr FA. Ordered dephosphorylation initiated by the selective proteolysis of cyclin B drives mitotic exit. eLife 2020; 9:e59885. [PMID: 32869743 PMCID: PMC7529458 DOI: 10.7554/elife.59885] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
APC/C-mediated proteolysis of cyclin B and securin promotes anaphase entry, inactivating CDK1 and permitting chromosome segregation, respectively. Reduction of CDK1 activity relieves inhibition of the CDK1-counteracting phosphatases PP1 and PP2A-B55, allowing wide-spread dephosphorylation of substrates. Meanwhile, continued APC/C activity promotes proteolysis of other mitotic regulators. Together, these activities orchestrate a complex series of events during mitotic exit. However, the relative importance of regulated proteolysis and dephosphorylation in dictating the order and timing of these events remains unclear. Using high temporal-resolution proteomics, we compare the relative extent of proteolysis and protein dephosphorylation. This reveals highly-selective rapid proteolysis of cyclin B, securin and geminin at the metaphase-anaphase transition, followed by slow proteolysis of other substrates. Dephosphorylation requires APC/C-dependent destruction of cyclin B and was resolved into PP1-dependent categories with unique sequence motifs. We conclude that dephosphorylation initiated by selective proteolysis of cyclin B drives the bulk of changes observed during mitotic exit.
Collapse
Affiliation(s)
- James Holder
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Shabaz Mohammed
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Francis A Barr
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
27
|
Houston J, Lara-Gonzalez P, Desai A. Rashomon at the kinetochore: Function(s) of the Mad1-cyclin B1 complex. J Cell Biol 2020; 219:151919. [PMID: 32614383 PMCID: PMC7401815 DOI: 10.1083/jcb.202006006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the film Rashomon, four witnesses describe seemingly contradictory views of one event. In a recent analogy, an interaction between the master mitotic regulator cyclin B1 and the spindle checkpoint component Mad1 was independently described by three groups who propose strikingly different functions for this interaction. Here, we summarize their findings and present a perspective on reconciling the different views.
Collapse
Affiliation(s)
- Jack Houston
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, CA.,Ludwig Institute for Cancer Research, San Diego Branch, La Jolla, CA
| | - Pablo Lara-Gonzalez
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, CA.,Ludwig Institute for Cancer Research, San Diego Branch, La Jolla, CA
| | - Arshad Desai
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, CA.,Ludwig Institute for Cancer Research, San Diego Branch, La Jolla, CA
| |
Collapse
|
28
|
Allan LA, Camacho Reis M, Ciossani G, Huis In 't Veld PJ, Wohlgemuth S, Kops GJ, Musacchio A, Saurin AT. Cyclin B1 scaffolds MAD1 at the kinetochore corona to activate the mitotic checkpoint. EMBO J 2020. [PMID: 32202322 DOI: 10.1525/embj.2019103180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
Cyclin B:CDK1 is the master kinase regulator of mitosis. We show here that, in addition to its kinase functions, mammalian Cyclin B also scaffolds a localised signalling pathway to help preserve genome stability. Cyclin B1 localises to an expanded region of the outer kinetochore, known as the corona, where it scaffolds the spindle assembly checkpoint (SAC) machinery by binding directly to MAD1. In vitro reconstitutions map the key binding interface to a few acidic residues in the N-terminal region of MAD1, and point mutations in this sequence abolish MAD1 corona localisation and weaken the SAC. Therefore, Cyclin B1 is the long-sought-after scaffold that links MAD1 to the corona, and this specific pool of MAD1 is needed to generate a robust SAC response. Robustness arises because Cyclin B1:MAD1 localisation loses dependence on MPS1 kinase after the corona has been established, ensuring that corona-localised MAD1 can still be phosphorylated when MPS1 activity is low. Therefore, this study explains how corona-MAD1 generates a robust SAC signal, and it reveals a scaffolding role for the key mitotic kinase, Cyclin B1:CDK1, which ultimately helps to inhibit its own degradation.
Collapse
Affiliation(s)
- Lindsey A Allan
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Magda Camacho Reis
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Giuseppe Ciossani
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Pim J Huis In 't Veld
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Sabine Wohlgemuth
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Geert Jpl Kops
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Adrian T Saurin
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| |
Collapse
|
29
|
Abstract
The spindle checkpoint protects against aneuploidy by ensuring that dividing cells only proceed with chromosome segregation once all kinetochores are stably attached to spindle microtubules. The checkpoint protein MAD1 localizes to the corona, a structural expansion of the kinetochore forming in the absence of microtubule attachment, but molecular mechanism or functional significance of this localization remains unknown. Recent results now show that cyclin B1 recruits MAD1 to the corona and that this MAD1 pool is required for robust checkpoint signaling.
Collapse
Affiliation(s)
- Carlos Conde
- Instituto de Investigação e Inovação em Saúde-i3S, Universidade do Porto, Porto, Portugal
| | - Reto Gassmann
- Instituto de Investigação e Inovação em Saúde-i3S, Universidade do Porto, Porto, Portugal
| |
Collapse
|
30
|
Allan LA, Camacho Reis M, Ciossani G, Huis in ‘t Veld PJ, Wohlgemuth S, Kops GJPL, Musacchio A, Saurin AT. Cyclin B1 scaffolds MAD1 at the kinetochore corona to activate the mitotic checkpoint. EMBO J 2020; 39:e103180. [PMID: 32202322 PMCID: PMC7298293 DOI: 10.15252/embj.2019103180] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/20/2020] [Accepted: 03/02/2020] [Indexed: 11/28/2022] Open
Abstract
Cyclin B:CDK1 is the master kinase regulator of mitosis. We show here that, in addition to its kinase functions, mammalian Cyclin B also scaffolds a localised signalling pathway to help preserve genome stability. Cyclin B1 localises to an expanded region of the outer kinetochore, known as the corona, where it scaffolds the spindle assembly checkpoint (SAC) machinery by binding directly to MAD1. In vitro reconstitutions map the key binding interface to a few acidic residues in the N-terminal region of MAD1, and point mutations in this sequence abolish MAD1 corona localisation and weaken the SAC. Therefore, Cyclin B1 is the long-sought-after scaffold that links MAD1 to the corona, and this specific pool of MAD1 is needed to generate a robust SAC response. Robustness arises because Cyclin B1:MAD1 localisation loses dependence on MPS1 kinase after the corona has been established, ensuring that corona-localised MAD1 can still be phosphorylated when MPS1 activity is low. Therefore, this study explains how corona-MAD1 generates a robust SAC signal, and it reveals a scaffolding role for the key mitotic kinase, Cyclin B1:CDK1, which ultimately helps to inhibit its own degradation.
Collapse
Affiliation(s)
- Lindsey A Allan
- Division of Cellular MedicineSchool of MedicineUniversity of DundeeDundeeUK
| | - Magda Camacho Reis
- Division of Cellular MedicineSchool of MedicineUniversity of DundeeDundeeUK
| | - Giuseppe Ciossani
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Pim J Huis in ‘t Veld
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Sabine Wohlgemuth
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Geert JPL Kops
- Oncode InstituteHubrecht Institute—KNAW and University Medical Centre UtrechtUtrechtThe Netherlands
| | - Andrea Musacchio
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Adrian T Saurin
- Division of Cellular MedicineSchool of MedicineUniversity of DundeeDundeeUK
| |
Collapse
|
31
|
Hégarat N, Crncec A, Suarez Peredo Rodriguez MF, Echegaray Iturra F, Gu Y, Busby O, Lang PF, Barr AR, Bakal C, Kanemaki MT, Lamond AI, Novak B, Ly T, Hochegger H. Cyclin A triggers Mitosis either via the Greatwall kinase pathway or Cyclin B. EMBO J 2020; 39:e104419. [PMID: 32350921 PMCID: PMC7265243 DOI: 10.15252/embj.2020104419] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/09/2020] [Accepted: 03/25/2020] [Indexed: 01/23/2023] Open
Abstract
Two mitotic cyclin types, cyclin A and B, exist in higher eukaryotes, but their specialised functions in mitosis are incompletely understood. Using degron tags for rapid inducible protein removal, we analyse how acute depletion of these proteins affects mitosis. Loss of cyclin A in G2-phase prevents mitotic entry. Cells lacking cyclin B can enter mitosis and phosphorylate most mitotic proteins, because of parallel PP2A:B55 phosphatase inactivation by Greatwall kinase. The final barrier to mitotic establishment corresponds to nuclear envelope breakdown, which requires a decisive shift in the balance of cyclin-dependent kinase Cdk1 and PP2A:B55 activity. Beyond this point, cyclin B/Cdk1 is essential for phosphorylation of a distinct subset of mitotic Cdk1 substrates that are essential to complete cell division. Our results identify how cyclin A, cyclin B and Greatwall kinase coordinate mitotic progression by increasing levels of Cdk1-dependent substrate phosphorylation.
Collapse
Affiliation(s)
- Nadia Hégarat
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | - Adrijana Crncec
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | | | | | - Yan Gu
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | - Oliver Busby
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | - Paul F Lang
- Department of BiochemistryUniversity of OxfordOxfordUK
| | - Alexis R Barr
- MRC London Institute of Medical ScienceImperial CollegeLondonUK
- Institute of Clinical SciencesFaculty of MedicineImperial CollegeLondonUK
| | - Chris Bakal
- Institute for Cancer ResearchChester Beatty LaboratoriesLondonUK
| | - Masato T Kanemaki
- National Institute of GeneticsResearch Organization of Information and Systems (ROIS)MishimaJapan
- Department of GeneticsSOKENDAI (The Graduate University of Advanced Studies)MishimaJapan
| | - Angus I Lamond
- Centre for Gene Regulation and ExpressionSchool of Life SciencesUniversity of DundeeDundeeUK
| | - Bela Novak
- Department of BiochemistryUniversity of OxfordOxfordUK
| | - Tony Ly
- Wellcome Trust Centre for Cell BiologyUniversity of EdinburghEdinburghUK
| | - Helfrid Hochegger
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| |
Collapse
|
32
|
Jackman M, Marcozzi C, Barbiero M, Pardo M, Yu L, Tyson AL, Choudhary JS, Pines J. Cyclin B1-Cdk1 facilitates MAD1 release from the nuclear pore to ensure a robust spindle checkpoint. J Cell Biol 2020; 219:e201907082. [PMID: 32236513 PMCID: PMC7265330 DOI: 10.1083/jcb.201907082] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/05/2020] [Accepted: 03/06/2020] [Indexed: 11/22/2022] Open
Abstract
How the cell rapidly and completely reorganizes its architecture when it divides is a problem that has fascinated researchers for almost 150 yr. We now know that the core regulatory machinery is highly conserved in eukaryotes, but how these multiple protein kinases, protein phosphatases, and ubiquitin ligases are coordinated in space and time to remodel the cell in a matter of minutes remains a major question. Cyclin B1-Cdk is the primary kinase that drives mitotic remodeling; here we show that it is targeted to the nuclear pore complex (NPC) by binding an acidic face of the kinetochore checkpoint protein, MAD1, where it coordinates NPC disassembly with kinetochore assembly. Localized cyclin B1-Cdk1 is needed for the proper release of MAD1 from the embrace of TPR at the nuclear pore so that it can be recruited to kinetochores before nuclear envelope breakdown to maintain genomic stability.
Collapse
|
33
|
Abstract
The goal of mitosis is to form two daughter cells each containing one copy of each mother cell chromosome, replicated in the previous S phase. To achieve this, sister chromatids held together back-to-back at their primary constriction, the centromere, have to interact with microtubules of the mitotic spindle so that each chromatid takes connections with microtubules emanating from opposite spindle poles (we will refer to this condition as bipolar attachment). Only once all replicated chromosomes have reached bipolar attachments can sister chromatids lose cohesion with each other, at the onset of anaphase, and move toward opposite spindle poles, being segregated into what will soon become the daughter cell nucleus. Prevention of errors in chromosome segregation is granted by a safeguard mechanism called Spindle Assembly Checkpoint (SAC). Until all chromosomes are bipolarly oriented at the equator of the mitotic spindle, the SAC prevents loss of sister chromatid cohesion, thus anaphase onset, and maintains the mitotic state by inhibiting inactivation of the major M phase promoting kinase, the cyclin B-cdk1 complex (Cdk1). Here, we review recent mechanistic insights about the circuitry that links Cdk1 to the SAC to ensure correct achievement of the goal of mitosis.
Collapse
Affiliation(s)
- Angela Flavia Serpico
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.,DMMBM, University of Naples "Federico II", Naples, 80131, Italy
| | - Domenico Grieco
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.,Department of Pharmacy, University of Naples "Federico II", Naples, 80131, Italy
| |
Collapse
|
34
|
Hayward D, Bancroft J, Mangat D, Alfonso-Pérez T, Dugdale S, McCarthy J, Barr FA, Gruneberg U. Checkpoint signaling and error correction require regulation of the MPS1 T-loop by PP2A-B56. J Cell Biol 2019; 218:3188-3199. [PMID: 31511308 PMCID: PMC6781431 DOI: 10.1083/jcb.201905026] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/19/2019] [Accepted: 08/06/2019] [Indexed: 01/16/2023] Open
Abstract
During mitosis, the formation of microtubule-kinetochore attachments is monitored by the serine/threonine kinase monopolar spindle 1 (MPS1). MPS1 is recruited to unattached kinetochores where it phosphorylates KNL1, BUB1, and MAD1 to initiate the spindle assembly checkpoint. This arrests the cell cycle until all kinetochores have been stably captured by microtubules. MPS1 also contributes to the error correction process rectifying incorrect kinetochore attachments. MPS1 activity at kinetochores requires autophosphorylation at multiple sites including threonine 676 in the activation segment or "T-loop." We now demonstrate that the BUBR1-bound pool of PP2A-B56 regulates MPS1 T-loop autophosphorylation and hence activation status in mammalian cells. Overriding this regulation using phosphomimetic mutations in the MPS1 T-loop to generate a constitutively active kinase results in a prolonged mitotic arrest with continuous turnover of microtubule-kinetochore attachments. Dynamic regulation of MPS1 catalytic activity by kinetochore-localized PP2A-B56 is thus critical for controlled MPS1 activity and timely cell cycle progression.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - James Bancroft
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | | | - Sholto Dugdale
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Julia McCarthy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Crncec A, Hochegger H. Triggering mitosis. FEBS Lett 2019; 593:2868-2888. [PMID: 31602636 DOI: 10.1002/1873-3468.13635] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/07/2019] [Accepted: 10/07/2019] [Indexed: 12/28/2022]
Abstract
Entry into mitosis is triggered by the activation of cyclin-dependent kinase 1 (Cdk1). This simple reaction rapidly and irreversibly sets the cell up for division. Even though the core step in triggering mitosis is so simple, the regulation of this cellular switch is highly complex, involving a large number of interconnected signalling cascades. We do have a detailed knowledge of most of the components of this network, but only a poor understanding of how they work together to create a precise and robust system that ensures that mitosis is triggered at the right time and in an orderly fashion. In this review, we will give an overview of the literature that describes the Cdk1 activation network and then address questions relating to the systems biology of this switch. How is the timing of the trigger controlled? How is mitosis insulated from interphase? What determines the sequence of events, following the initial trigger of Cdk1 activation? Which elements ensure robustness in the timing and execution of the switch? How has this system been adapted to the high levels of replication stress in cancer cells?
Collapse
Affiliation(s)
- Adrijana Crncec
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| | - Helfrid Hochegger
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| |
Collapse
|
36
|
Hayward D, Alfonso-Pérez T, Gruneberg U. Orchestration of the spindle assembly checkpoint by CDK1-cyclin B1. FEBS Lett 2019; 593:2889-2907. [PMID: 31469407 DOI: 10.1002/1873-3468.13591] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/01/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022]
Abstract
In mitosis, the spindle assembly checkpoint (SAC) monitors the formation of microtubule-kinetochore attachments during capture of chromosomes by the mitotic spindle. Spindle assembly is complete once there are no longer any unattached kinetochores. Here, we will discuss the mechanism and key components of spindle checkpoint signalling. Unattached kinetochores bind the principal spindle checkpoint kinase monopolar spindle 1 (MPS1). MPS1 triggers the recruitment of other spindle checkpoint proteins and the formation of a soluble inhibitor of anaphase, thus preventing exit from mitosis. On microtubule attachment, kinetochores become checkpoint silent due to the actions of PP2A-B56 and PP1. This SAC responsive period has to be coordinated with mitotic spindle formation to ensure timely mitotic exit and accurate chromosome segregation. We focus on the molecular mechanisms by which the SAC permissive state is created, describing a central role for CDK1-cyclin B1 and its counteracting phosphatase PP2A-B55. Furthermore, we discuss how CDK1-cyclin B1, through its interaction with MAD1, acts as an integral component of the SAC, and actively orchestrates checkpoint signalling and thus contributes to the faithful execution of mitosis.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, UK
| | | | | |
Collapse
|
37
|
Alfonso-Pérez T, Hayward D, Holder J, Gruneberg U, Barr FA. MAD1-dependent recruitment of CDK1-CCNB1 to kinetochores promotes spindle checkpoint signaling. J Cell Biol 2019; 218:1108-1117. [PMID: 30674583 PMCID: PMC6446853 DOI: 10.1083/jcb.201808015] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/19/2018] [Accepted: 01/07/2019] [Indexed: 12/22/2022] Open
Abstract
Cyclin B-dependent kinase (CDK1-CCNB1) promotes entry into mitosis. Additionally, it inhibits mitotic exit by activating the spindle checkpoint. This latter role is mediated through phosphorylation of the checkpoint kinase MPS1 and other spindle checkpoint proteins. We find that CDK1-CCNB1 localizes to unattached kinetochores and like MPS1 is lost from these structures upon microtubule attachment. This suggests that CDK1-CCNB1 is an integral component and not only an upstream regulator of the spindle checkpoint pathway. Complementary proteomic and cell biological analysis demonstrate that the spindle checkpoint protein MAD1 is one of the major components of CCNB1 complexes, and that CCNB1 is recruited to unattached kinetochores in an MPS1-dependent fashion through interaction with the first 100 amino acids of MAD1. This MPS1 and MAD1-dependent pool of CDK1-CCNB1 creates a positive feedback loop necessary for timely recruitment of MPS1 to kinetochores during mitotic entry and for sustained spindle checkpoint arrest. CDK1-CCNB1 is therefore an integral component of the spindle checkpoint, ensuring the fidelity of mitosis.
Collapse
Affiliation(s)
| | - Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - James Holder
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Recent Progress on the Localization of the Spindle Assembly Checkpoint Machinery to Kinetochores. Cells 2019; 8:cells8030278. [PMID: 30909555 PMCID: PMC6468716 DOI: 10.3390/cells8030278] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 12/14/2022] Open
Abstract
Faithful chromosome segregation during mitosis is crucial for maintaining genome stability. The spindle assembly checkpoint (SAC) is a surveillance mechanism that ensures accurate mitotic progression. Defective SAC signaling leads to premature sister chromatid separation and aneuploid daughter cells. Mechanistically, the SAC couples the kinetochore microtubule attachment status to the cell cycle progression machinery. In the presence of abnormal kinetochore microtubule attachments, the SAC prevents the metaphase-to-anaphase transition through a complex kinase-phosphatase signaling cascade which results in the correct balance of SAC components recruited to the kinetochore. The correct kinetochore localization of SAC proteins is a prerequisite for robust SAC signaling and, hence, accurate chromosome segregation. Here, we review recent progresses on the kinetochore recruitment of core SAC factors.
Collapse
|