1
|
Chen J, Wu S, He JJ, Liu YP, Deng ZY, Fang HK, Chen JF, Wei YL, She ZY. Kinesin-7 CENP-E mediates centrosome organization and spindle assembly to regulate chromosome alignment and genome stability. Cell Prolif 2025; 58:e13745. [PMID: 39266203 DOI: 10.1111/cpr.13745] [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: 05/22/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
Chromosome congression and alignment are essential for cell cycle progression and genomic stability. Kinesin-7 CENP-E, a plus-end-directed kinesin motor, is required for chromosome biorientation, congression and alignment in cell division. However, it remains unclear how chromosomes are aligned and segregated in the absence of CENP-E in mitosis. In this study, we utilize the CRISPR-Cas9 gene editing method and high-throughput screening to establish CENP-E knockout cell lines and reveal that CENP-E deletion results in defects in chromosome congression, alignment and segregation, which further promotes aneuploidy and genomic instability in mitosis. Both CENP-E inhibition and deletion lead to the dispersion of spindle poles, the formation of the multipolar spindle and spindle disorganization, which indicates that CENP-E is necessary for the organization and maintenance of spindle poles. In addition, CENP-E heterozygous deletion in spleen tissues also leads to the accumulation of dividing lymphocytes and cell cycle arrest in vivo. Furthermore, CENP-E deletion also disrupts the localization of key kinetochore proteins and triggers the activation of the spindle assembly checkpoint. In summary, our findings demonstrate that CENP-E promotes kinetochore-microtubule attachment and spindle pole organization to regulate chromosome alignment and spindle assembly checkpoint during cell division.
Collapse
Affiliation(s)
- Jie Chen
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Shan Wu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Jie-Jie He
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Yu-Peng Liu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Zhao-Yang Deng
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Han-Kai Fang
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Jian-Fan Chen
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Ya-Lan Wei
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| |
Collapse
|
2
|
Peng P, Zheng J, He K, Wang K, Wang L, Zheng X, Wu H, Yang Z, Zhang S, Zhao L. CENPE is a diagnostic and prognostic biomarker for cervical cancer. Heliyon 2024; 10:e40860. [PMID: 39759304 PMCID: PMC11698922 DOI: 10.1016/j.heliyon.2024.e40860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/26/2024] [Accepted: 11/30/2024] [Indexed: 01/07/2025] Open
Abstract
Cervical squamous cell carcinoma (CESC) is a common cancer in women. Despite advancements in early diagnosis through high-risk human papillomavirus (HPV) screening, challenges remain in predicting and treating the disease. Hence, the identification of novel biomarkers for prognosis and therapeutic targets is crucial. CENPE, a microtubule-end directed motor protein that accumulates during the G2 phase, is recognized for its involvement in promoting cancer growth and progression. However, its specific role in CESC remains unclear. This research investigated the expression of CENPE in CESC utilizing data from The Cancer Genome Atlas (TCGA), which was further validated through gene expression profiles, the Human Protein Atlas (HPA), and clinical data. The study utilized Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA), and immune infiltration analysis to elucidate the role of CENPE in CESC. Additionally, Protein-protein interaction (PPI) networks and competing endogenous RNA (CeRNA) networks involving CENPE and its differentially expressed genes were established. Furthermore, Kaplan-Meier survival analysis was conducted to evaluate the impact of CENPE on patient prognosis. Our study revealed an upregulation of CENPE expression in cervical cancer tissues, which promotes the progression of CESC through IL-6-mediated PI3K-Akt and MAPK signaling pathways. The significant associations with ACNG3, LY6H, and SLC6A7 suggest that CENPE may play a role in tumor growth and metastasis, potentially involving the nervous system. Moreover, the correlations with ARIH1, KDM1A, KDM5B, and NSD3 indicate that CENPE could be a promising target for drug development. Our analysis of the ROC curve demonstrated a high diagnostic accuracy of CENPE in CESC (AUC: 0.997, CI: 0.990-1.000). Subgroup analysis highlighted substantial effects in patients under 50 years old, those with a height under 160 cm, individuals in peri- and post-menopausal stages, and patients in clinical stages 1 and 4. Additionally, COX regression analysis indicated that older age, lower BMI, and higher CENPE expression are associated with decreased 1-year, 3-year, and 5-year survival rates. In conclusion, CENPE emerges as a crucial factor in the initiation and advancement of cervical cancer, showing potential as a novel target for therapeutic interventions.
Collapse
Affiliation(s)
- Peiqiang Peng
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Jingying Zheng
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, No.4026, Yatai Street, Changchun City, 130000, Jilin Province, China
| | - Kang He
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Kai Wang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Longyun Wang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Xufei Zheng
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Hao Wu
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Zhenning Yang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Shuang Zhang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Lijing Zhao
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| |
Collapse
|
3
|
Tang X, He Y, Tang Y, Chen K, Lin H, Liu B, Deng X. A kinetochore-associated kinesin-7 motor cooperates with BUB3.3 to regulate mitotic chromosome congression in Arabidopsis thaliana. NATURE PLANTS 2024; 10:1724-1736. [PMID: 39414927 DOI: 10.1038/s41477-024-01824-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 09/20/2024] [Indexed: 10/18/2024]
Abstract
Faithful genome partition during cell division relies on proper congression of chromosomes to the spindle equator before sister chromatid segregation. Here we uncover a kinesin-7 motor, kinetochore-associated kinesin 1 (KAK1), that is required for mitotic chromosome congression in Arabidopsis. KAK1 associates dynamically with kinetochores throughout mitosis. Loss of KAK1 results in severe defects in chromosome congression at metaphase, yet segregation errors at anaphase are rarely observed. KAK1 specifically interacts with the spindle assembly checkpoint protein BUB3.3 and both proteins show interdependent kinetochore localization. Chromosome misalignment in BUB3.3-depleted plants can be rescued by artificial tethering of KAK1 to kinetochores but not vice versa, demonstrating that KAK1 acts downstream of BUB3.3 to orchestrate microtubule-based chromosome transport at kinetochores. Moreover, we show that KAK1's motor activity is essential for driving chromosome congression to the metaphase plate. Thus, our findings reveal that plants have repurposed BUB3.3 to interface with a specialized kinesin adapted to integrate proper chromosome congression and checkpoint control through a distinct kinetochore design.
Collapse
Affiliation(s)
- Xiaoya Tang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Ying He
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yihang Tang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Keqi Chen
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Honghui Lin
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Bo Liu
- Department of Plant Biology, College of Biological Sciences, University of California, Davis, CA, USA
| | - Xingguang Deng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Zhang JL, Xu MF, Chen J, Wei YL, She ZY. Kinesin-7 CENP-E mediates chromosome alignment and spindle assembly checkpoint in meiosis I. Chromosoma 2024; 133:149-168. [PMID: 38456964 DOI: 10.1007/s00412-024-00818-w] [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: 03/27/2023] [Revised: 02/05/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
In eukaryotes, meiosis is the genetic basis for sexual reproduction, which is important for chromosome stability and species evolution. The defects in meiosis usually lead to chromosome aneuploidy, reduced gamete number, and genetic diseases, but the pathogenic mechanisms are not well clarified. Kinesin-7 CENP-E is a key regulator in chromosome alignment and spindle assembly checkpoint in cell division. However, the functions and mechanisms of CENP-E in male meiosis remain largely unknown. In this study, we have revealed that the CENP-E gene was highly expressed in the rat testis. CENP-E inhibition influences chromosome alignment and spindle organization in metaphase I spermatocytes. We have found that a portion of misaligned homologous chromosomes is located at the spindle poles after CENP-E inhibition, which further activates the spindle assembly checkpoint during the metaphase-to-anaphase transition in rat spermatocytes. Furthermore, CENP-E depletion leads to abnormal spermatogenesis, reduced sperm count, and abnormal sperm head structure. Our findings have elucidated that CENP-E is essential for homologous chromosome alignment and spindle assembly checkpoint in spermatocytes, which further contribute to chromosome stability and sperm cell quality during spermatogenesis.
Collapse
Affiliation(s)
- Jing-Lian Zhang
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Meng-Fei Xu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Jie Chen
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Ya-Lan Wei
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
5
|
Fang H, Zhang Y, Lin C, Sun Z, Wen W, Sheng H, Lin J. Primary microcephaly gene CENPE is a novel biomarker and potential therapeutic target for non-WNT/non-SHH medulloblastoma. Front Immunol 2023; 14:1227143. [PMID: 37593739 PMCID: PMC10427915 DOI: 10.3389/fimmu.2023.1227143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Background Non-WNT/non-SHH medulloblastoma (MB) is one of the subtypes with the highest genetic heterogeneity in MB, and its current treatment strategies have unsatisfactory results and significant side effects. As a member of the centromere protein (CENP) family, centromeric protein E (CENPE) is a microtubule plus-end-directed kinetochore protein. Heterozygous mutations in CENPE can leads to primary microcephaly syndrome. It has been reported that CENPE is upregulated in MB, but its role in MB development is still unknown. Methods We downloaded the relevant RNA seq data and matched clinical information from the GEO database. Bioinformatics analysis includes differential gene expression analysis, Kaplan-Meier survival analysis, nomogram analysis, ROC curve analysis, immune cell infiltration analysis, and gene function enrichment analysis. Moreover, the effects of CENPE expression on cell proliferation, cell cycle, and p53 signaling pathway of non-WNT/non-SHH MB were validated using CENPE specific siRNA in vitro experiments. Results Compared with normal tissues, CENPE was highly expressed in MB tissues and served as an independent prognostic factor for survival in non-WNT/non-SHH MB patients. The nomogram analysis and ROC curve further confirmed these findings. At the same time, immune cell infiltration analysis showed that CENPE may participate in the immune response and tumor microenvironment (TME) of non-WNT/non-SHH MB. In addition, gene enrichment analysis showed that CENPE was closely related to the cell cycle and p53 pathway in non-WNT/non-SHH MB. In vitro experimental validation showed that knockdown of CENPE inhibited cell proliferation by activating the p53 signaling pathway and blocking the cell cycle. Conclusion The expression of CENPE in non-WNT/non-SHH MB was positively correlated with poor prognosis. CENPE may affect tumor progression by regulating cell cycle, p53 pathway, and immune infiltration. Hence, CENPE is highly likely a novel biomarker and potential therapeutic target for non-WNT/non-SHH MB.
Collapse
Affiliation(s)
- Huangyi Fang
- Wenzhou Medical University, Wenzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yusong Zhang
- Wenzhou Medical University, Wenzhou, China
- Department of Surgery, The First People’s Hospital of Jiashan, Jiaxing, China
| | | | - Zhenkai Sun
- Wenzhou Medical University, Wenzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Wen
- Wenzhou Medical University, Wenzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hansong Sheng
- Wenzhou Medical University, Wenzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Lin
- Wenzhou Medical University, Wenzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Amin MA, Chakraborty M, Wallace DA, Varma D. Coordination between the Ndc80 complex and dynein is essential for microtubule plus-end capture by kinetochores during early mitosis. J Biol Chem 2023; 299:104711. [PMID: 37060995 PMCID: PMC10206188 DOI: 10.1016/j.jbc.2023.104711] [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: 10/27/2022] [Revised: 03/22/2023] [Accepted: 04/02/2023] [Indexed: 04/17/2023] Open
Abstract
Mitotic kinetochores are initially captured by dynamic microtubules via a "search-and-capture" mechanism. The microtubule motor, dynein, is critical for kinetochore capture as it has been shown to transport microtubule-attached chromosomes toward the spindle pole during prometaphase. The microtubule-binding nuclear division cycle 80 (Ndc80) complex that is recruited to kinetochores in prophase is known to play a central role in forming kinetochore-microtubule (kMT) attachments in metaphase. It is not yet clear, however, how Ndc80 contributes to initial kMT capture during prometaphase. Here, by combining CRISPR/Cas9-mediated knockout and RNAi technology with assays specific to study kMT capture, we show that mitotic cells lacking Ndc80 exhibit substantial defects in this function during prometaphase. Rescue experiments show that Ndc80 mutants deficient in microtubule-binding are unable to execute proper kMT capture. While cells inhibited of dynein alone are predominantly able to make initial kMT attachments, cells co-depleted of Ndc80 and dynein show severe defects in kMT capture. Further, we use an in vitro total internal reflection fluorescence microscopy assay to reconstitute microtubule capture events, which suggest that Ndc80 and dynein coordinate with each other for microtubule plus-end capture and that the phosphorylation status of Ndc80 is critical for productive kMT capture. A novel interaction between Ndc80 and dynein that we identify in prometaphase extracts might be critical for efficient plus-end capture. Thus, our studies, for the first time, identify a distinct event in the formation of initial kMT attachments, which is directly mediated by Ndc80 and in coordination with dynein is required for efficient kMT capture and chromosome alignment.
Collapse
Affiliation(s)
- Mohammed Abdullahel Amin
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | - Manas Chakraborty
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Destiny Ariel Wallace
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dileep Varma
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
7
|
Xie P. Determinant factors for residence time of kinesin motors at microtubule ends. J Biol Phys 2023; 49:77-93. [PMID: 36645568 PMCID: PMC9958224 DOI: 10.1007/s10867-022-09623-x] [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: 09/09/2022] [Accepted: 12/26/2022] [Indexed: 01/17/2023] Open
Abstract
Kinesins constitute a superfamily of microtubule (MT)-based motor proteins, which can perform diverse biological functions in cells such as transporting vesicle, regulating MT dynamics, and segregating chromosome. Some motors such as kinesin-1, kinesin-2, and kinesin-3 do the activity mainly on the MT lattice, while others such as kinesin-7 and kinesin-8 do the activity mainly at the MT plus end. To perform the different functions, it is required that the former motors can reside on the MT lattice for longer times than at the end, while the latter motors can reside at the MT plus end for long times. Here, a simple but general theory of the MT-end residence time of the kinesin motor is presented, with which the factors dictating the residence time are determined. The theory is further used to study specifically the MT-end residence times of Drosophila kinesin-1, kinesin-2/KIF3AB, kinesin-3/Unc104, kinesin-5/Eg5, kinesin-7/CENP-E, and kinesin-8/Kip3 motors, with the theoretical results being in agreement with the available experimental data.
Collapse
Affiliation(s)
- Ping Xie
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Science, Beijing, 100190, China.
| |
Collapse
|
8
|
Asymmetric chromatin retention and nuclear envelopes separate chromosomes in fused cells in vivo. Commun Biol 2022; 5:953. [PMID: 36123528 PMCID: PMC9485224 DOI: 10.1038/s42003-022-03874-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Abstract
Hybrid cells derived through fertilization or somatic cell fusion recognize and separate chromosomes of different origins. The underlying mechanisms are unknown but could prevent aneuploidy and tumor formation. Here, we acutely induce fusion between Drosophila neural stem cells (neuroblasts; NBs) and differentiating ganglion mother cells (GMCs) in vivo to define how epigenetically distinct chromatin is recognized and segregated. We find that NB-GMC hybrid cells align both endogenous (neuroblast-origin) and ectopic (GMC-origin) chromosomes at the metaphase plate through centrosome derived dual-spindles. Physical separation of endogenous and ectopic chromatin is achieved through asymmetric, microtubule-dependent chromatin retention in interphase and physical boundaries imposed by nuclear envelopes. The chromatin separation mechanisms described here could apply to the first zygotic division in insects, arthropods, and vertebrates or potentially inform biased chromatid segregation in stem cells. A hybrid fly cell model to test the separation of chromosomes of different origin. Neural stem cell (NB) - ganglion mother cell (GMC) hybrids align the respective chromosomes independently, supported by NB- or GMC-derived centrosomes and their spindles.
Collapse
|
9
|
She ZY, Xu MF, Jiang SY, Wei YL. Kinesin-7 CENP-E is essential for chromosome alignment and spindle assembly of mouse spermatocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119306. [PMID: 35680098 DOI: 10.1016/j.bbamcr.2022.119306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Genome stability depends on chromosome congression and alignment during cell division. Kinesin-7 CENP-E is critical for kinetochore-microtubule attachment and chromosome alignment, which contribute to genome stability in mitosis. However, the functions and mechanisms of CENP-E in the meiotic division of male spermatocytes remain largely unknown. In this study, by combining the use of chemical inhibitors, siRNA-mediated gene knockdown, immunohistochemistry, and high-resolution microscopy, we have found that CENP-E inhibition results in chromosome misalignment and metaphase arrest in dividing spermatocyte during meiosis. Strikingly, we have revealed that CENP-E regulates spindle organization in metaphase I spermatocytes and cultured GC-2 spd cells. CENP-E depletion leads to spindle elongation, chromosome misalignment, and chromosome instability in spermatocytes. Together, these findings indicate that CENP-E mediates the kinetochore recruitment of BubR1, spindle assembly checkpoint and chromosome alignment in dividing spermatocytes, which finally contribute to faithful chromosome segregation and chromosome stability in the male meiotic division.
Collapse
Affiliation(s)
- Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China; Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian 350122, China.
| | - Meng-Fei Xu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China; Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian 350122, China
| | - Sun-Ying Jiang
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China; Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian 350122, China
| | - Ya-Lan Wei
- Fujian Obstetrics and Gynecology Hospital, Fuzhou, Fujian 350011, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, China
| |
Collapse
|
10
|
Iegiani G, Di Cunto F, Pallavicini G. Inhibiting microcephaly genes as alternative to microtubule targeting agents to treat brain tumors. Cell Death Dis 2021; 12:956. [PMID: 34663805 PMCID: PMC8523548 DOI: 10.1038/s41419-021-04259-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/10/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
Medulloblastoma (MB) and gliomas are the most frequent high-grade brain tumors (HGBT) in children and adulthood, respectively. The general treatment for these tumors consists in surgery, followed by radiotherapy and chemotherapy. Despite the improvement in patient survival, these therapies are only partially effective, and many patients still die. In the last decades, microtubules have emerged as interesting molecular targets for HGBT, as various microtubule targeting agents (MTAs) have been developed and tested pre-clinically and clinically with encouraging results. Nevertheless, these treatments produce relevant side effects since they target microtubules in normal as well as in cancerous cells. A possible strategy to overcome this toxicity could be to target proteins that control microtubule dynamics but are required by HGBT cells much more than in normal cell types. The genes mutated in primary hereditary microcephaly (MCPH) are ubiquitously expressed in proliferating cells, but under normal conditions are selectively required during brain development, in neural progenitors. There is evidence that MB and glioma cells share molecular profiles with progenitors of cerebellar granules and of cortical radial glia cells, in which MCPH gene functions are fundamental. Moreover, several studies indicate that MCPH genes are required for HGBT expansion. Among the 25 known MCPH genes, we focus this review on KNL1, ASPM, CENPE, CITK and KIF14, which have been found to control microtubule stability during cell division. We summarize the current knowledge about the molecular basis of their interaction with microtubules. Moreover, we will discuss data that suggest these genes are promising candidates as HGBT-specific targets.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, 10043, Orbassano, Italy.
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, 10126, Turin, Italy.
| |
Collapse
|
11
|
Novais P, Silva PMA, Amorim I, Bousbaa H. Second-Generation Antimitotics in Cancer Clinical Trials. Pharmaceutics 2021; 13:1011. [PMID: 34371703 PMCID: PMC8309102 DOI: 10.3390/pharmaceutics13071011] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
Mitosis represents a promising target to block cancer cell proliferation. Classical antimitotics, mainly microtubule-targeting agents (MTAs), such as taxanes and vinca alkaloids, are amongst the most successful anticancer drugs. By disrupting microtubules, they activate the spindle assembly checkpoint (SAC), which induces a prolonged delay in mitosis, expected to induce cell death. However, resistance, toxicity, and slippage limit the MTA's effectiveness. With the desire to overcome some of the MTA's limitations, mitotic and SAC components have attracted great interest as promising microtubule-independent targets, leading to the so-called second-generation antimitotics (SGAs). The identification of inhibitors against most of these targets, and the promising outcomes achieved in preclinical assays, has sparked the interest of academia and industry. Many of these inhibitors have entered clinical trials; however, they exhibited limited efficacy as monotherapy, and failed to go beyond phase II trials. Combination therapies are emerging as promising strategies to give a second chance to these SGAs. Here, an updated view of the SGAs that reached clinical trials is here provided, together with future research directions, focusing on inhibitors that target the SAC components.
Collapse
Affiliation(s)
- Pedro Novais
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (P.N.); (P.M.A.S.)
- Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Patrícia M. A. Silva
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (P.N.); (P.M.A.S.)
| | - Isabel Amorim
- GreenUPorto (Sustainable Agrifood Production) Research Center, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal;
| | - Hassan Bousbaa
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (P.N.); (P.M.A.S.)
| |
Collapse
|
12
|
Pan J, Chao NX, Zhang YY, Huang TM, Chen CX, Qin QH, Guo JH, Huang RS, Luo GR. Upregulating KTN1 promotes Hepatocellular Carcinoma progression. J Cancer 2021; 12:4791-4809. [PMID: 34234850 PMCID: PMC8247380 DOI: 10.7150/jca.55570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 05/23/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) presents a common malignant tumor worldwide. Although kinectin 1 (KTN1) is the most frequently identified antigen in HCC tissues, the detailed roles of KTN1 in HCC remain unknown. This study seeks to clarify the expression status and clinical value of KTN1 in HCC and to explore the complicated biological functions of KTN1 and its underlying mechanisms. Methods: In-house reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression of KTN1 in HCC tissues. External gene microarrays and RNA-sequencing datasets were downloaded to confirm the expression patterns of KTN1. The prognostic ability of KTN1 in HCC was assessed by a Kaplan-Meier curve and a hazard ratio forest plot. The CRISPR/Cas9 gene-editing system was used to knock out KTN1 in Huh7 cells, which was verified by PCR-Sanger sequencing and western blotting. Assays of cell migration, invasion, viability, cell cycle, and apoptosis were conducted to explore the biological functions. RNA sequencing was performed to quantitatively analyze the functional deregulation in KTN1-knockout cells compared to Huh7-wild-type cells. Upregulated genes that co-expressed with KTN1 were identified from HCC tissues and were functionally annotated. Results: KTN1 expression was increased in HCC tissues (standardized mean difference [SMD] = 0.20 [0.04, 0.37]). High KTN1 expression was significantly correlated with poorer prognosis of HCC patients, and KTN1 may be an independent risk factor for HCC (pooled HRs = 1.31 [1.05, 1.64]). After KTN1-knockout, the viability, migration, and invasion ability of HCC cells were inhibited. The proportion of HCC cells in the G0-G1 phases increased after KTN1 knockout, which also elevated the apoptosis rates in HCC cells. Several cascades, including innate immune response, chemical carcinogenesis, and positive regulation of transcription by RNA polymerase II, were dramatically changed after KTN1 knockout. KTN1 primarily participated in the cell cycle, DNA replication, and microRNAs in cancer pathways in HCC tissues. Conclusion: Upregulation of KTN1 served as a promising prognosticator in HCC patients. KTN1 promotes the occurrence and deterioration of HCC by mediating cell survival, migration, invasion, cell cycle activation, and apoptotic inhibition. KTN1 may be a therapeutic target in HCC patients.
Collapse
Affiliation(s)
- Jian Pan
- Department of Human Anatomy, Guangxi Medical University.,Guangxi Colleges and Universities Key Laboratory of Human Development and Disease Research, Guangxi Medical University, Nanning, China
| | - Nai-Xia Chao
- Department of Biochemistry and Molecular Biology, Guangxi Medical University
| | - Yao-Yao Zhang
- Department of Histology and Embryology, Guangxi Medical University
| | - Tian-Ming Huang
- Department of Histology and Embryology, Guangxi Medical University
| | - Cheng-Xiao Chen
- The Ninth Affiliated Hospital of Guangxi Medical University, Guangxi Medical University
| | - Qiu-Hong Qin
- Jiang bin Hospital of Guangxi Zhuang Autonomous Region
| | | | - Rong-Shi Huang
- Department of Histology and Embryology, Guangxi Traditional Chinese Medical University
| | - Guo-Rong Luo
- Department of Histology and Embryology, Guangxi Medical University.,Guangxi Colleges and Universities Key Laboratory of Human Development and Disease Research, Guangxi Medical University, Nanning, China
| |
Collapse
|
13
|
Siskos N, Stylianopoulou E, Skavdis G, Grigoriou ME. Molecular Genetics of Microcephaly Primary Hereditary: An Overview. Brain Sci 2021; 11:brainsci11050581. [PMID: 33946187 PMCID: PMC8145766 DOI: 10.3390/brainsci11050581] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 11/16/2022] Open
Abstract
MicroCephaly Primary Hereditary (MCPH) is a rare congenital neurodevelopmental disorder characterized by a significant reduction of the occipitofrontal head circumference and mild to moderate mental disability. Patients have small brains, though with overall normal architecture; therefore, studying MCPH can reveal not only the pathological mechanisms leading to this condition, but also the mechanisms operating during normal development. MCPH is genetically heterogeneous, with 27 genes listed so far in the Online Mendelian Inheritance in Man (OMIM) database. In this review, we discuss the role of MCPH proteins and delineate the molecular mechanisms and common pathways in which they participate.
Collapse
|
14
|
Gama Braga L, Prifti DK, Garand C, Saini PK, Elowe S. A quantitative and semiautomated method for determining misaligned and lagging chromosomes during mitosis. Mol Biol Cell 2021; 32:880-891. [PMID: 33085580 PMCID: PMC8108530 DOI: 10.1091/mbc.e20-09-0585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 01/21/2023] Open
Abstract
Accurate chromosome alignment at metaphase facilitates the equal segregation of sister chromatids to each of the nascent daughter cells. Lack of proper metaphase alignment is an indicator of defective chromosome congression and aberrant kinetochore-microtubule attachments which in turn promotes chromosome missegregation and aneuploidy, hallmarks of cancer. Tools to sensitively, accurately, and quantitatively measure chromosome alignment at metaphase will facilitate understanding of the contribution of chromosome segregation errors to the development of aneuploidy. In this work, we have developed and validated a method based on analytical geometry to measure several indicators of chromosome misalignment. We generated semiautomated and flexible ImageJ2/Fiji pipelines to quantify kinetochore misalignment at metaphase plates as well as lagging chromosomes at anaphase. These tools will ultimately allow sensitive and systematic quantitation of these chromosome segregation defects in cells undergoing mitosis.
Collapse
Affiliation(s)
- Luciano Gama Braga
- Programme en Biologie cellulaire et moléculaire, Faculté de Médicine Université Laval, Québec, QC G1V 0A6, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de réproduction, santé de la mère et de l’enfant, Québec, QC G1V 4G2, Canada
- PROTEO-regroupement québécois de recherche sur la fonction, l’ingénierie et les applications des protéines, Québec, QC G1V 0A6, Canada
| | - Diogjena Katerina Prifti
- Programme en Biologie cellulaire et moléculaire, Faculté de Médicine Université Laval, Québec, QC G1V 0A6, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de réproduction, santé de la mère et de l’enfant, Québec, QC G1V 4G2, Canada
- PROTEO-regroupement québécois de recherche sur la fonction, l’ingénierie et les applications des protéines, Québec, QC G1V 0A6, Canada
| | - Chantal Garand
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de réproduction, santé de la mère et de l’enfant, Québec, QC G1V 4G2, Canada
| | - Pawan Kumar Saini
- Programme en Médicine moléculaire, Faculté de Médicine Université Laval, Québec, QC G1V 0A6, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de réproduction, santé de la mère et de l’enfant, Québec, QC G1V 4G2, Canada
- PROTEO-regroupement québécois de recherche sur la fonction, l’ingénierie et les applications des protéines, Québec, QC G1V 0A6, Canada
| | - Sabine Elowe
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de réproduction, santé de la mère et de l’enfant, Québec, QC G1V 4G2, Canada
- PROTEO-regroupement québécois de recherche sur la fonction, l’ingénierie et les applications des protéines, Québec, QC G1V 0A6, Canada
- Département de Pédiatire, Faculté de Médicine, Université Laval et le Centre de recherche sur le cancer de l’Université Laval, Québec, QC G1R 2J6, Canada
| |
Collapse
|
15
|
Iegiani G, Gai M, Di Cunto F, Pallavicini G. CENPE Inhibition Leads to Mitotic Catastrophe and DNA Damage in Medulloblastoma Cells. Cancers (Basel) 2021; 13:cancers13051028. [PMID: 33804489 PMCID: PMC7957796 DOI: 10.3390/cancers13051028] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Medulloblastoma (MB) is the most frequent brain tumor in children. The standard treatment consists in surgery, followed by radiotherapy and chemotherapy. These therapies are only partially effective, since many patients still die and those who survive suffer from neurological and endocrine disorders. Therefore, more effective therapies are needed. CENPE is a gene critical for normal proliferation and survival of neural progenitors. Since there is evidence that MB cells are very similar to neural progenitors, we hypothesized that CENPE could be an effective target for MB treatment. In MB cell lines, CENPE depletion induced defects in division and resulted in cell death. To consolidate CENPE as a target for MB treatment, we tested GSK923295, a specific inhibitor already in clinical trials for other cancer types. GSK923295 induced effects similar to CENPE depletion at low nM levels, supporting the idea that CENPE’s inhibition could be a viable strategy for MB treatment. Abstract Medulloblastoma (MB) is the most frequent brain tumor in children. The standard treatment consists in surgery, followed by radiotherapy and chemotherapy. These therapies are only partially effective since many patients still die and those who survive suffer from neurological and endocrine disorders. Therefore, more effective therapies are needed. Primary microcephaly (MCPH) is a rare disorder caused by mutations in 25 different genes. Centromere-associated protein E (CENPE) heterozygous mutations cause the MCPH13 syndrome. As for other MCPH genes, CENPE is required for normal proliferation and survival of neural progenitors. Since there is evidence that MB shares many molecular features with neural progenitors, we hypothesized that CENPE could be an effective target for MB treatment. In ONS-76 and DAOY cells, CENPE knockdown induced mitotic defects and apoptosis. Moreover, CENPE depletion induced endogenous DNA damage accumulation, activating TP53 or TP73 as well as cell death signaling pathways. To consolidate CENPE as a target for MB treatment, we tested GSK923295, an allosteric inhibitor already in clinical trial for other cancer types. GSK923295, induced effects similar to CENPE depletion with higher penetrance, at low nM levels, suggesting that CENPE’s inhibition could be a therapeutic strategy for MB treatment.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Turin, Italy;
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, 10126 Turin, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy;
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Turin, Italy;
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, 10126 Turin, Italy
- Correspondence: (F.D.C.); (G.P.)
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Turin, Italy;
- Department of Neuroscience ‘Rita Levi Montalcini’, University of Turin, 10126 Turin, Italy
- Correspondence: (F.D.C.); (G.P.)
| |
Collapse
|
16
|
Li J, Diao H, Guan X, Tian X. Kinesin Family Member C1 (KIFC1) Regulated by Centrosome Protein E (CENPE) Promotes Proliferation, Migration, and Epithelial-Mesenchymal Transition of Ovarian Cancer. Med Sci Monit 2020; 26:e927869. [PMID: 33361741 PMCID: PMC7780892 DOI: 10.12659/msm.927869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Centrosome amplification is recognized as a hallmark of cancer. Kinesin family member C1 (KIFC1), a centrosome-clustering molecule, is essential for the viability of extra centrosome-bearing cancer cells and may be the basis for the progression of ovarian cancer. However, its biological function and mechanism in ovarian cancer have not yet been studied. Material/Methods Quantitative reverse-transcription polymerase chain reaction was performed to detect the levels of KIFC1 and centrosome protein E (CENPE). Further, cell viability was analyzed with CCK-8 assay, and immunofluorescence was used to measure the expression of Ki67 and PCNA. Cell migration was analyzed with wound healing and transwell assays. Western blot analysis was performed to measure the expression of proteins in ovarian cancer cells. The relationship between KIFC1 and CENPE was investigated by performing co-immunoprecipitation. Results KIFC1 was upregulated in ovarian cancer cells, especially in SKOV3 cells. Additionally, we found that KIFC1 silencing in SKOV3 cells inhibited cell proliferation and downregulated the expression of Ki67 and PCNA. Further, the knockdown of KIFC1 suppressed cell migration and epithelial-mesenchymal transition (EMT) and regulated the expression of matrix metalloproteinase (MMP)2, MMP9, E-cadherin, N-cadherin, Snail, and ZEB1. Next, we found that KIFC1 bound to and positively regulated CENPE, a tumor promoter in certain human cancers. All the suppressive effects triggered by KIFC1 inhibition were reversed by CENPE overexpression. Conclusions KIFC1 contributed to cell proliferation, migration, and EMT via interacting with CENPE in ovarian cancer. KIFC1 might be a potential biomarker and therapeutic target in ovarian cancer patients.
Collapse
Affiliation(s)
- Jiangning Li
- Department of Gynecology, The Third People's Hospital of Dalian, Dalian, Liaoning, China (mainland)
| | - Haidan Diao
- Department of Gynecology, The Third People's Hospital of Dalian, Dalian, Liaoning, China (mainland)
| | - Xin Guan
- Department of Gynecology, The Third People's Hospital of Dalian, Dalian, Liaoning, China (mainland)
| | - Xiaofang Tian
- Department of Gynecology, The Third People's Hospital of Dalian, Dalian, Liaoning, China (mainland)
| |
Collapse
|
17
|
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: 1.8] [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
|
18
|
Montaudon E, Nikitorowicz-Buniak J, Sourd L, Morisset L, El Botty R, Huguet L, Dahmani A, Painsec P, Nemati F, Vacher S, Chemlali W, Masliah-Planchon J, Château-Joubert S, Rega C, Leal MF, Simigdala N, Pancholi S, Ribas R, Nicolas A, Meseure D, Vincent-Salomon A, Reyes C, Rapinat A, Gentien D, Larcher T, Bohec M, Baulande S, Bernard V, Decaudin D, Coussy F, Le Romancer M, Dutertre G, Tariq Z, Cottu P, Driouch K, Bièche I, Martin LA, Marangoni E. PLK1 inhibition exhibits strong anti-tumoral activity in CCND1-driven breast cancer metastases with acquired palbociclib resistance. Nat Commun 2020; 11:4053. [PMID: 32792481 PMCID: PMC7426966 DOI: 10.1038/s41467-020-17697-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 07/16/2020] [Indexed: 02/08/2023] Open
Abstract
A significant proportion of patients with oestrogen receptor (ER) positive breast cancers (BC) develop resistance to endocrine treatments (ET) and relapse with metastatic disease. Here we perform whole exome sequencing and gene expression analysis of matched primary breast tumours and bone metastasis-derived patient-derived xenografts (PDX). Transcriptomic analyses reveal enrichment of the G2/M checkpoint and up-regulation of Polo-like kinase 1 (PLK1) in PDX. PLK1 inhibition results in tumour shrinkage in highly proliferating CCND1-driven PDX, including different RB-positive PDX with acquired palbociclib resistance. Mechanistic studies in endocrine resistant cell lines, suggest an ER-independent function of PLK1 in regulating cell proliferation. Finally, in two independent clinical cohorts of ER positive BC, we find a strong association between high expression of PLK1 and a shorter metastases-free survival and poor response to anastrozole. In conclusion, our findings support clinical development of PLK1 inhibitors in patients with advanced CCND1-driven BC, including patients progressing on palbociclib treatment.
Collapse
Affiliation(s)
- Elodie Montaudon
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | | | - Laura Sourd
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Ludivine Morisset
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Rania El Botty
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Léa Huguet
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Ahmed Dahmani
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Pierre Painsec
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Fariba Nemati
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie, Paris, France
| | | | | | | | - Camilla Rega
- Institute of Cancer Research, 123 Old Brompton Road, SW7 3RP, London, UK
| | | | - Nikiana Simigdala
- Institute of Cancer Research, 123 Old Brompton Road, SW7 3RP, London, UK
| | - Sunil Pancholi
- Institute of Cancer Research, 123 Old Brompton Road, SW7 3RP, London, UK
| | - Ricardo Ribas
- Institute of Cancer Research, 123 Old Brompton Road, SW7 3RP, London, UK
| | - André Nicolas
- Department of Pathology, Institut Curie, Paris, France
| | | | | | - Cécile Reyes
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Audrey Rapinat
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - David Gentien
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
| | - Thibaut Larcher
- INRA, APEX-PAnTher, Oniris, Rue De La Géraudière Cedex 3, 44322, Nantes, France
| | - Mylène Bohec
- Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, Paris, France
| | - Sylvain Baulande
- Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, Paris, France
| | | | - Didier Decaudin
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Florence Coussy
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Muriel Le Romancer
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, 28 Rue Laennec, 69000, Lyon, France
| | | | - Zakia Tariq
- Department of Genetics, Institut Curie, Paris, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, Paris, France
| | | | - Ivan Bièche
- Department of Genetics, Institut Curie, Paris, France
| | - Lesley-Ann Martin
- Institute of Cancer Research, 123 Old Brompton Road, SW7 3RP, London, UK
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, 26 Rue d'Ulm, 75005, Paris, France.
| |
Collapse
|
19
|
Jang J, Engleka KA, Liu F, Li L, Song G, Epstein JA, Li D. An Engineered Mouse to Identify Proliferating Cells and Their Derivatives. Front Cell Dev Biol 2020; 8:388. [PMID: 32523954 PMCID: PMC7261916 DOI: 10.3389/fcell.2020.00388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/29/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Cell proliferation is a fundamental event during development, disease, and regeneration. Effectively tracking and quantifying proliferating cells and their derivatives is critical for addressing many research questions. Cell cycle expression such as for Ki67, proliferating cell nuclear antigen (PCNA), or aurora kinase B (Aurkb), or measurement of 5-bromo-2'-deoxyuridine (BrdU) or 3H-thymidine incorporation have been widely used to assess and quantify cell proliferation. These are powerful tools for detecting actively proliferating cells, but they do not identify cell populations derived from proliferating progenitors over time. AIMS We developed a new mouse tool for lineage tracing of proliferating cells by targeting the Aurkb allele. RESULTS In quiescent cells or cells arrested at G1/S, little or no Aurkb mRNA is detectable. In cycling cells, Aurkb transcripts are detectable at G2 and become undetectable by telophase. These findings suggest that Aurkb transcription is restricted to proliferating cells and is tightly coupled to cell proliferation. Accordingly, we generated an Aurkb ER Cre/+ mouse by targeting a tamoxifen inducible Cre cassette into the start codon of Aurkb. We find that the Aurkb ER Cre/+ mouse faithfully labels proliferating cells in developing embryos and regenerative adult tissues such as intestine but does not label quiescent cells such as post-mitotic neurons. CONCLUSION The Aurkb ER Cre/+ mouse faithfully labels proliferating cells and their derivatives in developing embryos and regenerative adult tissues. This new mouse tool provides a novel genetic tracing capability for studying tissue proliferation and regeneration.
Collapse
Affiliation(s)
- Jihyun Jang
- Department of Surgery, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kurt A. Engleka
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Feiyan Liu
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Li Li
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guang Song
- Department of Surgery, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jonathan A. Epstein
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Deqiang Li
- Department of Surgery, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|