1
|
Shi Y, Pan Q, Chen W, Xie L, Tang S, Yang Z, Zhang M, Yin D, Lin L, Liao JY. Pan-cancer oncogenic properties and therapeutic potential of SF3B4. Cancer Gene Ther 2025:10.1038/s41417-025-00910-y. [PMID: 40394232 DOI: 10.1038/s41417-025-00910-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 05/22/2025]
Abstract
Splicing factor 3B (SF3B) subunit 4 (SF3B4), an SF3B complex component essential for spliceosome assembly and accurate splicing, plays a major role in cancer development. However, the precise mechanism through which SF3B4 contributes to tumor growth remains unclear. Here, we demonstrate that SF3B4 is strongly expressed in patients with various cancer types and correlated with their survival. By using hepatocellular carcinoma (HCC) as a model, we reveal that SF3B4's interactions with and regulatory influence on the checkpoint protein BUB1 are essential for appropriate cancer cell mitosis and proliferation. Our results thus demonstrate the roles of SF3B4 as both a cell-cycle regulator and an oncogenic factor in HCC, highlighting its potential as a pan-cancer therapeutic target and diagnostic biomarker.
Collapse
Affiliation(s)
- Yanmei Shi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Qimei Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Wenli Chen
- Center for Bioresources and Drug Discovery and School of Biosciences and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Limin Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Shiru Tang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Zhizhi Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Man Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China.
| | - Jian-You Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China.
| |
Collapse
|
2
|
Chapagai D, Strebhardt K, Wyatt MD, McInnes C. Structural regulation of PLK1 activity: implications for cell cycle function and drug discovery. Cancer Gene Ther 2025:10.1038/s41417-025-00907-7. [PMID: 40379873 DOI: 10.1038/s41417-025-00907-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/19/2025]
Abstract
Polo Like Kinase 1 (PLK1), a key regulator of mitosis whose overexpression is often associated with poor survival rates in cancer, continues to be widely investigated as an oncology drug target with clinical trials evaluating second and third generation inhibitors. In addition to the conserved N-terminal kinase domain (KD), a unique characteristic of the Polo-Like kinase family is the C-terminal polo-box domain (PBD). The PBD contains a phosphopeptide binding site that recognizes substrates primed by other kinases and furthermore is responsible for subcellular localization of PLK1 to specific sites in the nucleus including centrosomes and kinetochores. Another role of the PBD is its regulatory ability through domain-domain interactions with the KD to maintain an autoinhibited state of PLK1. Insights into post translational modifications and the PBD - KD domain-domain association have been obtained and show that key events in PLK1 regulation include phosphosubstrate binding, T210 phosphorylation and engagement with the Bora protein. These can induce an open and active conformation where the domain-domain inhibitory interactions no longer dominate. Further regulatory events recently described include the interchange between monomeric and dimeric forms, which can also serve to inhibit or activate PLK1 during the cell cycle. Different oligomeric forms of PLK1, existing as homodimers and heterodimers with PLK2, have been identified and likely play context dependent roles. This review provides an overview of recent information describing structural and mechanistic insights into inhibition of PLK1 and the temporal and spatial requirements of its activation and regulation. It also covers recent insights into the conformational regulation of other members of the Polo-Like kinase family. The implications of the conformational regulation of PLK1 with respect to cell cycle function and drug discovery are significant and are therefore discussed in detail.
Collapse
Affiliation(s)
- Danda Chapagai
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, 02129, USA
| | - Klaus Strebhardt
- Department of Gynecology, University Hospital, Goethe University, Theodor-Stern-Kai 7-9, Frankfurt am Main, 60596, Germany
| | - Michael D Wyatt
- Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, 29208, USA
| | - Campbell McInnes
- Drug Discovery and Biomedical Sciences, University of South Carolina, 715 Sumter Street, Columbia, 29208, USA.
| |
Collapse
|
3
|
Schweighofer J, Mulay B, Hoffmann I, Vogt D, Pesenti ME, Musacchio A. Interactions with multiple inner kinetochore proteins determine mitotic localization of FACT. J Cell Biol 2025; 224:e202412042. [PMID: 40094435 PMCID: PMC11912937 DOI: 10.1083/jcb.202412042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
The FAcilitates Chromatin Transcription (FACT) complex is a dimeric histone chaperone that operates on chromatin during transcription and replication. FACT also interacts with a specialized centromeric nucleosome containing the histone H3 variant centromere protein A (CENP-A) and with CENP-TW, two subunits of the constitutive centromere-associated network (CCAN), a 16-protein complex associated with CENP-A. The significance of these interactions remains elusive. Here, we show that FACT has multiple additional binding sites on CCAN. The interaction with CCAN is strongly stimulated by casein kinase II phosphorylation of FACT. Mitotic localization of FACT to kinetochores is strictly dependent on specific CCAN subcomplexes. Conversely, CENP-TW requires FACT for stable localization. Unexpectedly, we also find that DNA readily displaces FACT from CCAN, supporting the speculation that FACT becomes recruited through a pool of CCAN that is not stably integrated into chromatin. Collectively, our results point to a potential role of FACT in chaperoning CCAN during transcription or in the stabilization of CCAN at the centromere during the cell cycle.
Collapse
Affiliation(s)
- Julia Schweighofer
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Bhagyashree Mulay
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Ingrid Hoffmann
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Doro Vogt
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Marion E. Pesenti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
- Max Planck School Matter to Life, Heidelberg, Germany
| |
Collapse
|
4
|
Wu RZ, Sun QQ, Fu Y, Yu HN, Liu WY, Wu YH, Zhang H, Pan YL, Rui X. Fatty acid metabolism-derived prognostic model for lung adenocarcinoma: unraveling the link to survival and immune response. Front Immunol 2025; 16:1507845. [PMID: 40181976 PMCID: PMC11965909 DOI: 10.3389/fimmu.2025.1507845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is one of the most common malignant tumors globally, characterized by poor prognosis and high mortality. Abnormal fatty acid metabolism plays a crucial role in LUAD progression. This study aims to develop a prognostic model based on fatty acid metabolism to improve the overall prognosis of LUAD. Materials and methods Bioinformatics analyses were performed using TCGA and GEO datasets, supplemented by cell experiments. A total of 309 fatty acid metabolism-related genes were identified from MsigDB. Differentially expressed genes were analyzed using the 'limma' R package. A prognostic model was constructed using LASSO regression and validated with survival analyses via the 'survminer', 'survival', and 'pROC' R packages. The analysis included somatic mutations, tumor mutation burden, clinical correlations, stemness analysis, cytokine correlations, and enrichment analysis. Protein interaction networks were constructed using STRING and Cytoscape, while immune cell infiltration and immunotherapy responses were evaluated with the 'oncoPredict' R package. Results were validated through cell experiments and immunohistochemistry staining of lung tissues. Results We identified 125 differentially expressed genes related to fatty acid metabolism, with 33 genes significantly associated with prognosis. Patients in the high-risk group had poorer overall survival and progression-free survival, and the risk score correlated with gender, N stage, clinical stage, and T stage. The risk score was also associated with cancer stem cells, with a significantly higher mRNAsi index in the high-risk group. Additionally, the risk score correlated with various cytokine expressions and showed significant enrichment in cell cycle pathways. Key genes like CDK1 were highly expressed in LUAD cell lines and validated in clinical samples. The low-risk group showed better responses to immune checkpoint inhibitors, with the risk score correlating with immune checkpoint gene expression. Conclusion This study successfully established a novel prognostic model based on fatty acid metabolism, which provides valuable insights for the treatment of LUAD.
Collapse
Affiliation(s)
- Rui-Ze Wu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Qian-Qian Sun
- School of Public Health, Harbin Medical University, Harbin, China
| | - Yao Fu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Han-Nong Yu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Wei-Yang Liu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Yong-Hui Wu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Han Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu-Lin Pan
- School of Public Health, Harbin Medical University, Harbin, China
| | - Xin Rui
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
5
|
Shen Z, Adams K, Moreno R, Lera R, Kaufman E, Lang JD, Burkard M. Polo-like kinase 1 maintains transcription and chromosomal accessibility during mitosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637959. [PMID: 39990329 PMCID: PMC11844518 DOI: 10.1101/2025.02.12.637959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Transcription persists at low levels in mitotic cells and plays essential roles in mitotic fidelity and chromosomal dynamics. However, the detailed regulatory network of mitotic transcription remains largely unresolved. Here, we report the novel role of Polo-like kinase 1 (Plk1) in maintaining mitotic transcription. Using 5-ethynyl uridine (5-EU) labeling of nascent RNAs, we found that Plk1 inhibition leads to significant downregulation of nascent transcription in prometaphase cells. Chromatin-localized Plk1 activity is required for transcription regulation and mitotic fidelity. Plk1 sustains global chromosomal accessibility in mitosis, especially at promoter and transcription start site (promoter-TSS) regions, facilitating transcription factor binding and ensuring proper transcriptional activity. We identified SMC4, a common subunit of condensin I and II, as a potential Plk1 substrate. Plk1 activity is fundamental to these processes across non-transformed and transformed cell lines, underscoring its critical role in cell cycle regulation. This study elucidates a novel regulatory mechanism of global mitotic transcription, advancing our understanding of cell cycle control. Significance Statement Cells retain a low level of transcription during mitosis, while the regulatory network and specific contributions of mitotic transcription are not well understood.We identify Polo-like kinase 1 (Plk1) as a novel regulator of mitotic transcription, crucial for chromosome condensation, genome accessibility, and maintaining mitotic fidelity.This study enhances our understanding of Plk1's multifaceted role in mitotic progression, advancing cell cycle regulation knowledge, and informing new cancer therapies' development.
Collapse
|
6
|
Kozgunova E. Recent advances in plant kinetochore research. Front Cell Dev Biol 2025; 12:1510019. [PMID: 39911184 PMCID: PMC11794483 DOI: 10.3389/fcell.2024.1510019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Faithful chromosome segregation is crucial for cell division in eukaryotes, facilitated by the kinetochore, a multi-subunit protein complex that connects chromosomes to the spindle microtubules. Recent research has significantly advanced our understanding of kinetochore function in plants, including surprising findings about spindle assembly checkpoint, the composition of the inner kinetochore and unique kinetochore arrangement in holocentric Cuscuta species. Additionally, some kinetochore proteins in plants have been implicated in roles beyond chromosome segregation, such as cytokinesis regulation and involvement in developmental processes. This review summarizes recent insights into plant kinetochore biology, compares plant kinetochores with those of animals and fungi, and highlights key open questions and potential future directions in the field.
Collapse
Affiliation(s)
- Elena Kozgunova
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
7
|
Kong W, Hara M, Tokunaga Y, Okumura K, Hirano Y, Miao J, Takenoshita Y, Hashimoto M, Sasaki H, Fujimori T, Wakabayashi Y, Fukagawa T. CENP-C-Mis12 complex establishes a regulatory loop through Aurora B for chromosome segregation. Life Sci Alliance 2025; 8:e202402927. [PMID: 39433344 PMCID: PMC11494776 DOI: 10.26508/lsa.202402927] [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: 07/04/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Establishing the correct kinetochore-microtubule attachment is crucial for faithful chromosome segregation. The kinetochore has various regulatory mechanisms for establishing correct bipolar attachment. However, how the regulations are coupled is not fully understood. Here, we demonstrate a regulatory loop between the kinetochore protein CENP-C and Aurora B kinase, which is critical for the error correction of kinetochore-microtubule attachment. This regulatory loop is mediated through the binding of CENP-C to the outer kinetochore Mis12 complex (Mis12C). Although the Mis12C-binding region of CENP-C is dispensable for mouse development and proliferation in human RPE-1 cells, those cells lacking this region display increased mitotic defects. The CENP-C-Mis12C interaction facilitates the centromeric recruitment of Aurora B and the mitotic error correction in human cells. Given that Aurora B reinforces the CENP-C-Mis12C interaction, our findings reveal a positive regulatory loop between Aurora B recruitment and the CENP-C-Mis12C interaction, which ensures chromosome biorientation for accurate chromosome segregation.
Collapse
Affiliation(s)
- Weixia Kong
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Masatoshi Hara
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yurika Tokunaga
- Division of Experimental Animal Research, Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Kazuhiro Okumura
- Division of Experimental Animal Research, Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yasuhiro Hirano
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Jiahang Miao
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | | | - Masakazu Hashimoto
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Cell Science, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hiroshi Sasaki
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Yuichi Wakabayashi
- Division of Experimental Animal Research, Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
8
|
Zou C, Zhang Y, Liu C, Li Y, Lin C, Chen H, Hou J, Gao G, Liu Z, Yan Q, Su W. Identification of CENPM as a key gene driving adrenocortical carcinoma metastasis via physical interaction with immune checkpoint ligand FGL1. Clin Transl Med 2025; 15:e70182. [PMID: 39778025 PMCID: PMC11707433 DOI: 10.1002/ctm2.70182] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Distant metastasis occurs in the majority of adrenocortical carcinoma (ACC), leading to an extremely poor prognosis. However, the key genes driving ACC metastasis remain unclear. METHODS Weighted gene co-expression network analysis (WGCNA) and functional enrichment analysis were conducted to identify ACC metastasis-related genes. Data from RNA-seq and microarray were analyzed to reveal correlations of the CENPM gene with cancer, metastasis, and survival in ACC. Immunohistochemistry was used to assess CENPM protein expression. The impact of CENPM on metastasis behaviour was verified in ACC (H295R and SW-13) cells and xenograft NPG mice. DIA quantitative proteomics analysis, western blot, immunofluorescence, and co-immunoprecipitation assay were performed to identify the downstream target of CENPM. RESULTS Among the 12 035 analyzed genes, 363 genes were related to ACC metastasis and CENPM was identified as the hub gene. CENPM was upregulated in ACC samples and associated with metastasis and poor prognosis. Knockdown of CENPM inhibited proliferation, invasion, and migration of ACC cells and suppressed liver metastasis in xenograft NPG mice. Collagen-containing extracellular matrix signalling was primarily downregulated when CENPM was knocked down. FGL1, important components of ECM signalling and immune checkpoint ligand of LAG3, were downregulated following CENPM silence, overexpressed in human advanced ACC samples, and colocalized with CENPM. Physical interaction between CENPM and FGL1 was identified. Overexpression of FGL1 rescued migration and invasion of CENPM knockdown ACC cells. CONCLUSIONS CENPM is a key gene in driving ACC metastasis. CENPM promotes ACC metastasis through physical interaction with the immune checkpoint ligand FGL1. CENPM can be used as a new prognostic biomarker and therapeutic target for metastatic ACC. HIGHLIGHTS CENPM is the key gene that drives ACC metastasis, and a robust biomarker for ACC prognosis. Silencing CENPM impedes ACC metastasis in vitro and in vivo by physical interaction with immune checkpoint ligand FGL1. FGL1 is overexpressed in ACC and promotes ACC metastasis.
Collapse
Affiliation(s)
- Cunru Zou
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Yu Zhang
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Chengyue Liu
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Yaxin Li
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Congjie Lin
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Hao Chen
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Jiangping Hou
- Department of OphthalmologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Guojun Gao
- Department of Urology SurgeryAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Zheng Liu
- Department of Urology SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Qiupeng Yan
- Department of Teaching and Research Section of Introduction to Basic MedicineSchool of Basic MedicineShandong Second Medical UniversityWeifangChina
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher EducationShandong Second Medical UniversityWeifangChina
| | - Wenxia Su
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| |
Collapse
|
9
|
Black EM, Ramírez Parrado CA, Trier I, Li W, Joo YK, Pichurin J, Liu Y, Kabeche L. Chk2 sustains PLK1 activity in mitosis to ensure proper chromosome segregation. Nat Commun 2024; 15:10782. [PMID: 39737931 PMCID: PMC11685634 DOI: 10.1038/s41467-024-54922-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Polo-like kinase 1 (PLK1) protects against genome instability by ensuring timely and accurate mitotic cell division, and its activity is tightly regulated throughout the cell cycle. Although the pathways that initially activate PLK1 in G2 are well-characterized, the factors that directly regulate mitotic PLK1 remain poorly understood. Here, we identify that human PLK1 activity is sustained by the DNA damage response kinase Checkpoint kinase 2 (Chk2) in mitosis. Chk2 directly phosphorylates PLK1 T210, a residue on its T-loop whose phosphorylation is essential for full PLK1 kinase activity. Loss of Chk2-dependent PLK1 activity causes increased mitotic errors, including chromosome misalignment, chromosome missegregation, and cytokinetic defects. Moreover, Chk2 deficiency increases sensitivity to PLK1 inhibitors, suggesting that Chk2 status may be an informative biomarker for PLK1 inhibitor efficacy. This work demonstrates that Chk2 sustains mitotic PLK1 activity and protects genome stability through discrete functions in interphase DNA damage repair and mitotic chromosome segregation.
Collapse
Affiliation(s)
- Elizabeth M Black
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Carlos Andrés Ramírez Parrado
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Isabelle Trier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Wenxue Li
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Yoon Ki Joo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Jennifer Pichurin
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Yansheng Liu
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA.
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA.
| |
Collapse
|
10
|
Chen YC, Kilic E, Wang E, Rossman W, Suzuki A. CENcyclopedia: Dynamic Landscape of Kinetochore Architecture Throughout the Cell Cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627000. [PMID: 39677682 PMCID: PMC11643120 DOI: 10.1101/2024.12.05.627000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The kinetochore, an intricate macromolecular protein complex located on chromosomes, plays a pivotal role in orchestrating chromosome segregation. It functions as a versatile platform for microtubule assembly, diligently monitors microtubule binding fidelity, and acts as a force coupler. Comprising over 100 distinct proteins, many of which exist in multiple copies, the kinetochore's composition dynamically changes throughout the cell cycle, responding to specific timing and conditions. This dynamicity is important for establishing functional kinetochores, yet the regulatory mechanisms of these dynamics have largely remained elusive. In this study, we employed advanced quantitative immunofluorescence techniques to meticulously chart the dynamics of kinetochore protein levels across the cell cycle. These findings offer a comprehensive view of the dynamic landscape of kinetochore architecture, shedding light on the detailed mechanisms of microtubule interaction and the nuanced characteristics of kinetochore proteins. This study significantly advances our understanding of the molecular coordination underlying chromosome segregation.
Collapse
Affiliation(s)
- Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ece Kilic
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Evelyn Wang
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Will Rossman
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
11
|
Fesquet D, Rabeharivelo G, van Dijk J, Prigent C, Morin N, Rouquier S. CCDC69 maintains genome integrity by regulating KIF2C/MCAK depolymerase activity and the stability of the chromosomal passenger complex. Sci Rep 2024; 14:30401. [PMID: 39638803 PMCID: PMC11621681 DOI: 10.1038/s41598-024-81022-9] [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: 09/10/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024] Open
Abstract
Accurate genome inheritance during cell division relies on a complex chromosome segregation mechanism. This process occurs once all the kinetochores of sister chromatids are attached to microtubules emanating from the opposite poles of the mitotic spindle. To control the precision of this mechanism, the Chromosome Passenger Complex (CPC) actively identifies and corrects improper microtubule attachments. The depolymerase activity of the kinesin KIF2C/MCAK at the kinetochores is involved in this process. CCDC69 is a poorly characterized protein, primarily identified as a regulator of central spindle assembly, whose overexpression prompts rapid microtubule depolymerization. Here, we show that CCDC69 is a cell-cycle regulated protein belonging to the Microtubule-associated Tumor Suppressor (MTUS) superfamily, and even slight deregulation of its expression induces severe early mitotic phenotypes. Myristoylation anchors CCDC69 at the plasma membrane, thus protecting microtubule network integrity. We found that CCDC69 microtubule depolymerization activity relies on KIF2C, with a fraction of CCDC69 localizing to the centromere. Importantly, we demonstrated that CCDC69 regulates the stability of the CPC by safeguarding its members from degradation during mitosis. In summary, our findings underscore CCDC69's essential role as a mitotic regulator, which is crucial for maintaining the fidelity of chromosome segregation.
Collapse
Affiliation(s)
- Didier Fesquet
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Gabriel Rabeharivelo
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Juliette van Dijk
- CRBM CNRS UMR 5237, Equipe Centrosome Cil Et Pathologies, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Claude Prigent
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Nathalie Morin
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Sylvie Rouquier
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France.
| |
Collapse
|
12
|
Fischer J, Erkner E, Radszuweit P, Hentrich T, Keppeler H, Korkmaz F, Schulze-Hentrich J, Fitzel R, Lengerke C, Schneidawind D, Schneidawind C. Only Infant MLL-Rearranged Leukemia Is Susceptible to an Inhibition of Polo-like Kinase 1 (PLK-1) by Volasertib. Int J Mol Sci 2024; 25:12760. [PMID: 39684470 DOI: 10.3390/ijms252312760] [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: 09/29/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
MLL-rearranged (MLLr) leukemia is characterized by a poor prognosis. Depending on the cell of origin, it differs in the aggressiveness and therapy response. For instance, in adults, volasertib blocking Polo-like kinase 1 (PLK-1) exhibited limited success. Otherwise, PLK-1 characterizes an infant MLLr signature, indicating potential sensitivity. By using our CRISPR/Cas9 MLLr model in CD34+ cells from human cord blood (huCB) and bone marrow (huBM) mimicking the infant and adult patient diseases, we were able to shed light on this phenomenon. The PLK-1 mRNA level was significantly increased in our huCB compared to the huBM model, which was underpinned by analyzing infant and adult MLLr leukemia patients. Importantly, the expression levels correlated with a functional response. Volasertib induced a significant dose-dependent decrease in proliferation and cell cycle arrest, most pronounced in the infant model. Mechanistically, upon volasertib treatment, we uncovered negative feedback only in the huBM model by compensatory upregulation of PLK-1 and related genes like AURKA involved in mitosis. Importantly, the poor response could be overcome by a combinatorial strategy with alisertib, an Aurora kinase A inhibitor. Our study emphasizes the importance of considering the cell of origin in therapeutic decision-making and provides the rationale for evaluating volasertib and alisertib in MLLr leukemia.
Collapse
Affiliation(s)
- Jacqueline Fischer
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
| | - Estelle Erkner
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
| | - Pia Radszuweit
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Thomas Hentrich
- Department of Genetics/Epigenetics, Faculty NT, Saarland University, 66123 Saarbruecken, Germany
| | - Hildegard Keppeler
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
| | - Fulya Korkmaz
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
| | - Julia Schulze-Hentrich
- Department of Genetics/Epigenetics, Faculty NT, Saarland University, 66123 Saarbruecken, Germany
| | - Rahel Fitzel
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
| | - Claudia Lengerke
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
| | - Dominik Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Corina Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, 72074 Tuebingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
13
|
Gao J, Zhu L, Yue X, Jing S, Tang S, Lee I, Qian Y. Golgin45 assists mitosis via its nuclear localization sequence. Biochem Biophys Res Commun 2024; 735:150845. [PMID: 39442450 DOI: 10.1016/j.bbrc.2024.150845] [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: 10/08/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
In mammalian cells, the Golgi apparatus undergoes fragmentation for its correct partition into two daughter cells during mitosis. Several Golgi structural proteins have been demonstrated to regulate Golgi disassembly/reassembly and spindle formation. However, it is largely unknown whether Golgi proteins mediate other major events in mitosis. Here, we report that Golgin45, a Golgi tethering protein, participates in recruiting PLK1 to the kinetochores. Upon entry into mitosis, Golgin45 binds PLK1 and a nuclear import protein, importin β2. Enriched RanGTP at kinetochores in prometaphase and metaphase sequesters importin β2 from Golgin45 and liberates Golgin45-PLK1 complex, which then gets further delivered to the kinetochores by Golgin45-KNL1 interaction. R375A mutation in Golgin45 that specifically disrupts Golgin45-importin β2 interaction impairs PLK1 localization to the kinetochores, leading to mitotic arrest. Our findings reveal a novel role of a golgin tether protein in mediating Ran-dependent PLK1 enrichment on the kinetochores for proper progression of mitosis.
Collapse
Affiliation(s)
- Jingkai Gao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shuocheng Tang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
14
|
Bel Borja L, Taylor SJP, Soubigou F, Pelisch F. CENP-C-targeted PLK-1 regulates kinetochore function in C. elegans embryos. J Cell Sci 2024; 137:jcs262327. [PMID: 39355896 PMCID: PMC11634037 DOI: 10.1242/jcs.262327] [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: 06/06/2024] [Accepted: 09/20/2024] [Indexed: 10/03/2024] Open
Abstract
Polo-like kinase 1 (PLK-1) is present in centrosomes, the nuclear envelope and kinetochores and plays a significant role in meiosis and mitosis. PLK-1 depletion or inhibition has severe consequences for spindle assembly, spindle assembly checkpoint (SAC) activation, chromosome segregation and cytokinesis. BUB-1 targets PLK-1 to the outer kinetochore and, in mammals, the inner kinetochore PLK1 targeting is mediated by the constitutive centromere associated network (CCAN). BUB-1-targeted PLK-1 plays a key role in SAC activation and has a SAC-independent role through targeting CDC-20. In contrast, whether there is a specific, non-redundant role for inner kinetochore targeted PLK-1 is unknown. Here, we used the Caenorhabditis elegans embryo to study the role of inner kinetochore PLK-1. We found that CENP-C, the sole CCAN component in C. elegans and other species, targets PLK-1 to the inner kinetochore during prometaphase and metaphase. Disruption of the CENP-C-PLK-1 interaction leads to an imbalance in kinetochore components and a defect in chromosome congression, without affecting CDC-20 recruitment. These findings indicate that PLK-1 kinetochore recruitment by CENP-C has at least partially distinct functions from outer kinetochore PLK-1, providing a platform for a better understanding of the different roles played by PLK-1 during mitosis.
Collapse
Affiliation(s)
- Laura Bel Borja
- Molecular, Cellular and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Samuel J. P. Taylor
- Molecular, Cellular and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Flavie Soubigou
- Molecular, Cellular and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Federico Pelisch
- Molecular, Cellular and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
15
|
Song C, Zhang M, Kruse T, Møller MH, López-Méndez B, Zhang Y, Zhai Y, Wang Y, Lei T, Kettenbach AN, Nilsson J, Zhang G. Self-priming of Plk1 binding to BubR1 ensures accurate mitotic progression. Commun Biol 2024; 7:1473. [PMID: 39516273 PMCID: PMC11549336 DOI: 10.1038/s42003-024-07205-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Plk1 is a key mitotic kinase that localizes to distinct subcellular structures to promote accurate mitotic progression. Plk1 recruitment depends on direct interaction between polo-box domain (PBD) on Plk1 and PBD binding motif (PBD BM) on the interactors. However, recent study showed that PBD BM alone is not enough for stable binding between CENP-U and Plk1 highlighting the complexity of the interaction which warrants further investigation. An important interactor for Plk1 during mitosis is the checkpoint protein BubR1. Plk1 bound to BubR1 via PBD interaction with pT620 phosphorylates BubR1 S676/T680 to promote BubR1-PP2A/B56 interaction. The BubR1-PP2A/B56 complex counteracts the destablizing effect on kinetochore-microtubule attachments by mitotic kinases to promote mitotic progression. Here we show that Plk1 phosphorylates T600/T608 on BubR1 and the double phosphorylation is critical for BubR1-Plk1 interaction. A similar mechanism for Plk1-Bub1 interaction also exists indicating a general principle for Plk1 kinetochore recruitment through self-priming. Mechanistically preventing BubR1 T600/T608 phosphorylation impairs chromosome congression and checkpoint silencing by reducing Plk1 and PP2A/B56 binding to BubR1. Increasing the binding affinity towards Plk1 and PP2A/B56 in BubR1 through protein engineering bypasses the requirement of T600/T608 phosphorylation for mitotic progression. These results reveal a new layer of regulation for accurate mitotic progression.
Collapse
Affiliation(s)
- Chunlin Song
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Mingzhe Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Thomas Kruse
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mads Harder Møller
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blanca López-Méndez
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yuqing Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yujing Zhai
- School of Public Health, Qingdao University, Qingdao, China
| | - Ying Wang
- School of Public Health, Qingdao University, Qingdao, China
| | - Tingting Lei
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH, 03755, USA
| | - Jakob Nilsson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gang Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.
| |
Collapse
|
16
|
Stewart MG, Carrier JS, Zahm JA, Harrison SC, Miller MP. A coordinated kinase and phosphatase network regulates Stu2 recruitment to yeast kinetochores. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621564. [PMID: 39553986 PMCID: PMC11566010 DOI: 10.1101/2024.11.01.621564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Cells coordinate diverse events at anaphase onset, including separase activation, cohesin cleavage, chromosome separation, and spindle reorganization. Regulation of the XMAP215 family member and microtubule polymerase, Stu2, at the metaphase-anaphase transition determines a specific redistribution from kinetochores to spindle microtubules. We show that cells modulate Stu2 kinetochore-microtubule localization by Polo-like kinase1/Cdc5-mediated phosphorylation of T866, near the Stu2 C-terminus, thereby promoting dissociation from the kinetochore Ndc80 complex. Cdk/Cdc28 likely primes Cdc5:Stu2 interaction. Cdc28 activity is also required for Stu2 nuclear import. PP2ACdc55 actively opposes Cdc5 activity on Stu2T866 during metaphase. This counter-regulation allows for switchlike redistribution of Stu2pT866 at anaphase onset when separase inhibits PP2ACdc55. Blocking Stu2T866 phosphorylation disrupts anaphase spindle progression, and we infer that PP2ACdc55 regulates the mitotic spindle by dephosphorylating Stu2 and other MAPs. These data support a model in which increased phosphorylation at anaphase onset results from phosphatase inhibition and point to a larger regulatory network that facilitates rapid cytoskeletal modulation required for anaphase spindle maintenance.
Collapse
Affiliation(s)
- Michael G. Stewart
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Joseph S. Carrier
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Jacob A. Zahm
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Howard Hughes Medical Institute, Boston, United States
| | - Stephen C. Harrison
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Howard Hughes Medical Institute, Boston, United States
| | - Matthew P. Miller
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| |
Collapse
|
17
|
Lim WM, Chew WX, Esposito Verza A, Pesenti M, Musacchio A, Surrey T. Regulation of minimal spindle midzone organization by mitotic kinases. Nat Commun 2024; 15:9213. [PMID: 39472429 PMCID: PMC11522559 DOI: 10.1038/s41467-024-53500-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024] Open
Abstract
During cell division, the microtubule cytoskeleton undergoes dramatic cell cycle-driven reorganizations of its architecture. Coordinated by changes in the phosphorylation patterns of a multitude of microtubule associated proteins, the mitotic spindle first self-assembles to capture the chromosomes and then reorganizes in anaphase as the chromosomes are segregated. A key protein for this reorganization is PRC1 which is differentially phosphorylated by the mitotic kinases CDK1 and PLK1. How the phosphorylation state of PRC1 orchestrates spindle reorganization is not understood mechanistically. Here, we reconstitute in vitro the transition between metaphase and anaphase-like microtubule architectures triggered by the changes in PRC1 phosphorylation. We find that whereas PLK1 regulates its own recruitment by PRC1, CDK1 controls the affinity of PRC1 for antiparallel microtubule binding. Dephosphorylation of CDK1-phosphorylated PRC1 is required and sufficient to trigger the reorganization of a minimal anaphase midzone in the presence of the midzone length controlling kinesin KIF4A. These results demonstrate how phosphorylation-controlled affinity changes regulate the architecture of active microtubule networks, providing new insight into the mechanistic underpinnings of the cell cycle-driven reorganization of the central spindle during mitosis.
Collapse
Affiliation(s)
- Wei Ming Lim
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Carrer del Dr. Aiguader 88, Barcelona, Spain
| | - Wei-Xiang Chew
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Carrer del Dr. Aiguader 88, Barcelona, Spain
| | - Arianna Esposito Verza
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Marion Pesenti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Max Planck School Matter to Life, Heidelberg, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Thomas Surrey
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Carrer del Dr. Aiguader 88, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig de Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
18
|
Qi N, Wang B, Xing W, Ge F, Liu J. The protective role of quercetin against copper-induced female reproductive toxicity: Insights from transcriptome analysis. Food Chem Toxicol 2024; 192:114934. [PMID: 39151877 DOI: 10.1016/j.fct.2024.114934] [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: 03/26/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Quercetin has been shown to mitigate the cytotoxic effects of heavy metals. While copper is an essential trace element for bodily functions, excessive intake has been linked to impaired female reproductive function. Transcriptome analysis was employed to identify genes that are differentially expressed in response to high copper and were validated through qRT-PCR and western blotting. ATP content and Tunel were used to identify the damage of mitochondrial and cell apoptosis. PPI analysis revealed that MKI67, TOPII, ASPM, CASP3, PLK1, and TTK are central proteins within the network. Additionally, exposure to elevated levels of copper resulted in the dysregulation of 86 genes associated with mitochondria. Conversely, treatment with quercetin (QUE) in combination with high copper led to the normalization of 42 mitochondria-related genes previously affected by high copper levels. Furthermore, CuSO4 decreases ATP content and induces cell apoptosis, which can be reversed by QUE. Results suggest that elevated copper levels could lead to oxidative stress and apoptosis by inducing mitochondrial damage, while QUE has the potential to mitigate these effects, ultimately safeguarding granulosa cells and halting the progression of cell death. This study provides novel insights into the molecular pathways involved in female reproductive toxicity caused by excessive copper exposure.
Collapse
Affiliation(s)
- Nannan Qi
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| | - Binbin Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| | - Wenwen Xing
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| | - Fangcai Ge
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| | - Jiying Liu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| |
Collapse
|
19
|
Dwivedi D, Meraldi P. Balancing Plk1 activity levels: The secret of synchrony between the cell and the centrosome cycle. Bioessays 2024; 46:e2400048. [PMID: 39128131 DOI: 10.1002/bies.202400048] [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: 02/29/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
The accuracy of cell division requires precise regulation of the cellular machinery governing DNA/genome duplication, ensuring its equal distribution among the daughter cells. The control of the centrosome cycle is crucial for the formation of a bipolar spindle, ensuring error-free segregation of the genome. The cell and centrosome cycles operate in close synchrony along similar principles. Both require a single duplication round in every cell cycle, and both are controlled by the activity of key protein kinases. Nevertheless, our comprehension of the precise cellular mechanisms and critical regulators synchronizing these two cycles remains poorly defined. Here, we present our hypothesis that the spatiotemporal regulation of a dynamic equilibrium of mitotic kinases activities forms a molecular clock that governs the synchronous progression of both the cell and the centrosome cycles.
Collapse
Affiliation(s)
- Devashish Dwivedi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
20
|
Devillers R, Dos Santos A, Destombes Q, Laplante M, Elowe S. Recent insights into the causes and consequences of chromosome mis-segregation. Oncogene 2024; 43:3139-3150. [PMID: 39278989 DOI: 10.1038/s41388-024-03163-5] [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: 07/13/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
Mitotic cells face the challenging task of ensuring accurate and equal segregation of their duplicated, condensed chromosomes between the nascent daughter cells. Errors in the process result in chromosome missegregation, a significant consequence of which is the emergence of aneuploidy-characterized by an imbalance in chromosome number-and the associated phenomenon of chromosome instability (CIN). Aneuploidy and CIN are common features of cancer, which leverages them to promote genome heterogeneity and plasticity, thereby facilitating rapid tumor evolution. Recent research has provided insights into how mitotic errors shape cancer genomes by inducing both numerical and structural chromosomal changes that drive tumor initiation and progression. In this review, we survey recent findings regarding the mitotic causes and consequences of aneuploidy. We discuss new findings into the types of chromosome segregation errors that lead to aneuploidy and novel pathways that protect genome integrity during mitosis. Finally, we describe new developments in our understanding of the immediate consequences of chromosome mis-segregation on the genome stability of daughter cells.
Collapse
Affiliation(s)
- Romain Devillers
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Alexsandro Dos Santos
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Quentin Destombes
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Mathieu Laplante
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Sabine Elowe
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada.
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada.
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada.
- Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec City, QC, Canada.
| |
Collapse
|
21
|
Conti D, Verza AE, Pesenti ME, Cmentowski V, Vetter IR, Pan D, Musacchio A. Role of protein kinase PLK1 in the epigenetic maintenance of centromeres. Science 2024; 385:1091-1097. [PMID: 39236163 DOI: 10.1126/science.ado5178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/30/2024] [Indexed: 09/07/2024]
Abstract
The centromere, a chromosome locus defined by the histone H3-like protein centromeric protein A (CENP-A), promotes assembly of the kinetochore to bind microtubules during cell division. Centromere maintenance requires CENP-A to be actively replenished by dedicated protein machinery in the early G1 phase of the cell cycle to compensate for its dilution after DNA replication. Cyclin-dependent kinases (CDKs) limit CENP-A deposition to once per cell cycle and function as negative regulators outside of early G1. Antithetically, Polo-like kinase 1 (PLK1) promotes CENP-A deposition in early G1, but the molecular details of this process are still unknown. We reveal here a phosphorylation network that recruits PLK1 to the deposition machinery to control a conformational switch required for licensing the CENP-A deposition reaction. Our findings clarify how PLK1 contributes to the epigenetic maintenance of centromeres.
Collapse
Affiliation(s)
- Duccio Conti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Arianna Esposito Verza
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Marion E Pesenti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Verena Cmentowski
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Ingrid R Vetter
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Dongqing Pan
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, 45141 Essen, Germany
| |
Collapse
|
22
|
Li W, Hao Y. Polo-Like Kinase 1 and DNA Damage Response. DNA Cell Biol 2024; 43:430-437. [PMID: 38959179 DOI: 10.1089/dna.2024.0018] [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] [Indexed: 07/05/2024] Open
Abstract
Polo-like kinase 1 (Plk1), an evolutionarily conserved serine/threonine protein kinase, is a key regulator involved in the mitotic process of the cell cycle. Mounting evidence suggests that Plk1 is also involved in a variety of nonmitotic events, including the DNA damage response, DNA replication, cytokinesis, embryonic development, apoptosis, and immune regulation. The DNA damage response (DDR) includes activation of the DNA checkpoint, DNA damage recovery, DNA repair, and apoptosis. Plk1 is not only an important target of the G2/M DNA damage checkpoint but also negatively regulates the G2/M checkpoint commander Ataxia telangiectasia-mutated (ATM), promotes G2/M phase checkpoint recovery, and regulates homologous recombination repair by interacting with Rad51 and BRCA1, the key factors of homologous recombination repair. This article briefly reviews the function of Plk1 in response to DNA damage.
Collapse
Affiliation(s)
- Wei Li
- Laboratory of Nuclear and Radiation Damage, Characteristic Medical Center, PLA Rocket Force, Beijing, China
- Department of Disease Prevention and Control, Characteristic Medical Center, PLA Rocket Force, Beijing, China
| | - Yongjian Hao
- Department of Disease Prevention and Control, Characteristic Medical Center, PLA Rocket Force, Beijing, China
| |
Collapse
|
23
|
Bellah SF, Xiong F, Dou Z, Yang F, Liu X, Yao X, Gao X, Zhang L. PLK1 phosphorylation of ZW10 guides accurate chromosome segregation in mitosis. J Mol Cell Biol 2024; 16:mjae008. [PMID: 38402459 PMCID: PMC11328731 DOI: 10.1093/jmcb/mjae008] [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: 09/03/2023] [Revised: 11/09/2023] [Accepted: 02/23/2024] [Indexed: 02/26/2024] Open
Abstract
Stable transmission of genetic information during cell division requires faithful chromosome segregation. Mounting evidence has demonstrated that polo-like kinase 1 (PLK1) dynamics at kinetochores control correct kinetochore-microtubule attachments and subsequent silencing of the spindle assembly checkpoint. However, the mechanisms underlying PLK1-mediated silencing of the spindle checkpoint remain elusive. Here, we identified a regulatory mechanism by which PLK1-elicited zeste white 10 (ZW10) phosphorylation regulates spindle checkpoint silencing in mitosis. ZW10 is a cognate substrate of PLK1, and the phosphorylation of ZW10 at Ser12 enables dynamic ZW10-Zwint1 interactions. Inhibition of ZW10 phosphorylation resulted in misaligned chromosomes, while persistent expression of phospho-mimicking ZW10 mutant caused premature anaphase, in which sister chromatids entangled as cells entered anaphase. These findings reveal the previously uncharacterized PLK1-ZW10 interaction through which dynamic phosphorylation of ZW10 fine-tunes accurate chromosome segregation in mitosis.
Collapse
Affiliation(s)
- Sm Faysal Bellah
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| | - Fangyuan Xiong
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| | - Zhen Dou
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Xinjiao Gao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Liangyu Zhang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| |
Collapse
|
24
|
Chen S, Sun Q, Yao B, Ren Y. The Molecular Mechanism of Aurora-B Regulating Kinetochore-Microtubule Attachment in Mitosis and Oocyte Meiosis. Cytogenet Genome Res 2024; 164:69-77. [PMID: 39068909 DOI: 10.1159/000540588] [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: 01/08/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Aurora kinase B (Aurora-B), a member of the chromosomal passenger complex, is involved in correcting kinetochore-microtubule (KT-MT) attachment errors and regulating sister chromatid condensation and cytoplasmic division during mitosis. SUMMARY However, few reviews have discussed its mechanism in oocyte meiosis and the differences between its role in mitosis and meiosis. Therefore, in this review, we summarize the localization, recruitment, activation, and functions of Aurora-B in mitosis and oocyte meiosis. The accurate regulation of Aurora-B is essential for ensuring accurate chromosomal segregation and correct KT-MT attachments. Aurora-B regulates the stability of KT-MT attachments by competing with cyclin-dependent kinase 1 to control the phosphorylation of the SILK and RVSF motifs on kinetochore scaffold 1 and by competing with protein phosphatase 1 to influence the phosphorylation of NDC80 which is the substrate of Aurora-B. In addition, Aurora-B regulates the spindle assembly checkpoint by promoting the recruitment and activation of mitotic arrest deficient 2. KEY MESSAGES This review provides a theoretical foundation for elucidating the mechanism of cell division and understanding oocyte chromosomal aneuploidy.
Collapse
Affiliation(s)
- Shanshan Chen
- Department of Histology and Embryology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, China,
| | - Qiqi Sun
- Department of Histology and Embryology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Bo Yao
- Department of Histology and Embryology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Yanping Ren
- Department of Histology and Embryology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| |
Collapse
|
25
|
Yang H, Chen XW, Song XJ, Du HY, Si FC. Baitouweng decoction suppresses growth of esophageal carcinoma cells through miR-495-3p/BUB1/STAT3 axis. World J Gastrointest Oncol 2024; 16:3193-3210. [PMID: 39072160 PMCID: PMC11271792 DOI: 10.4251/wjgo.v16.i7.3193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/29/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Esophageal carcinoma (EC) is one of the most prevalent cancers in human populations worldwide. Baitouweng decoction is one of the most important Chinese medicine formulas, with the potential to treat cancer. AIM To investigate the role and mechanism of Baitouweng decoction on EC cells. METHODS Differentially expressed genes (DEGs) in EC tissues and normal tissues were screened by the cDNA microarray technique and by bioinformatics methods. The target genes of microRNAs were predicted based on the TargetScan database and verified by dual luciferase gene reporter assay. We used Baitouweng decoction to intervene EC cells, and detected the activity of EC9706 and KYSE150 cells by the MTT method. Cell cycle and apoptosis were measured by flow cytometry. The expression of BUB1 mRNA and miR-495-3p was measured by qRT-PCR. The protein levels of BUB1, STAT3, p-STAT3, CCNB1, CDK1, Bax, Caspase3, and Caspase9 were measured by Western blot analysis. The migration and invasion abilities of the cells were measured by wound-healing assay and Transwell invasion assay, respectively. RESULTS DEGs identified are involved in biological processes, signaling pathways, and network construction, which are mainly related to mitosis. BUB1 was the key hub gene, and it is also a target gene of miR-495-3p. Baitouweng decoction could upregulate miR-495-3p and inhibit BUB1 expression. In vitro experiments showed that Baitouweng decoction significantly inhibited the migration and invasion of EC cells and induced apoptosis and G2/M phase arrest. After treatment with Baitouweng decoction, the expression of Bax, Caspase 3, and Caspase 9 in EC cells increased significantly, while the expression of BUB1, CCNB1, and CDK1 decreased significantly. Moreover, the STAT3 signaling pathway may play an important role in this process. CONCLUSION Baitouweng decoction has a significant inhibitory effect on EC cell growth. BUB1 is a potential therapeutic target for EC. Further analysis showed that Baitouweng decoction may inhibit the growth of EC cells by upregulating miR-495-3p targeting the BUB1-mediated STAT3 signal pathway.
Collapse
Affiliation(s)
- Hui Yang
- Henan Key Laboratory of Traditional Chinese Medicine Syndrome and Prescription in Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Xiao-Wei Chen
- Henan Key Laboratory of Traditional Chinese Medicine Syndrome and Prescription in Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Xue-Jie Song
- Henan Key Laboratory of Traditional Chinese Medicine Syndrome and Prescription in Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hai-Yang Du
- Henan Key Laboratory of Traditional Chinese Medicine Syndrome and Prescription in Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Fu-Chun Si
- Henan Key Laboratory of Traditional Chinese Medicine Syndrome and Prescription in Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine School, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| |
Collapse
|
26
|
Lin C, Xiong J, Chen Y, Zheng H, Li M. Overexpression of CENPU promotes cancer growth and metastasis and is associated with poor survival in patients with nasopharyngeal carcinoma. Transl Cancer Res 2024; 13:2812-2824. [PMID: 38988917 PMCID: PMC11231766 DOI: 10.21037/tcr-23-2395] [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: 12/30/2023] [Accepted: 04/28/2024] [Indexed: 07/12/2024]
Abstract
Background Centromere protein U (CENPU) is key for mitosis in the carcinogenesis of cancers. However, the roles of CENPU have not been inspected in nasopharyngeal carcinoma (NPC). Thus, we aimed to explore the functions and mechanisms of CENPU in NPC. Methods Expression of CENPU was evaluated by real-time quantitative polymerase chain reaction, western blotting and immunohistochemistry. The biological functions of CENPU were evaluated in vitro and in vivo. Gene chip analysis, ingenuity pathway analysis, and coimmunoprecipitation experiments were used to explore the mechanisms of CENPU. Results CENPU was highly expressed in NPC. High expression of CENPU was associated with advanced tumor, node and metastasis (TNM) stage and poor overall survival. Cox regression analysis demonstrated that CENPU expression was an independent prognostic factor in NPC. Knockdown of CENPU inhibited proliferation and migration in vitro and in vivo. Knockdown of CENPU upregulated dual specificity phosphatase 6 (DUSP6) expression. The expression of CNEPU was inversely correlated with the expression of DUSP6 in NPC tissues. Mechanistic studies confirmed that CENPU increased the activation of the ERK1/2 and p38 signaling pathways by suppressing the expression of DUSP6. Conclusions CENPU acts as an oncogene in NPC by interacting with DUSP6, and may represent a promising prognostic biomarker for patients with NPC.
Collapse
Affiliation(s)
- Cheng Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jiani Xiong
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yuebing Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Huiping Zheng
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Meifang Li
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
27
|
Yan L, Yuan X, Liu M, Chen Q, Zhang M, Xu J, Zeng LH, Zhang L, Huang J, Lu W, He X, Yan H, Wang F. A non-canonical role of the inner kinetochore in regulating sister-chromatid cohesion at centromeres. EMBO J 2024; 43:2424-2452. [PMID: 38714893 PMCID: PMC11182772 DOI: 10.1038/s44318-024-00104-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/27/2024] [Accepted: 04/12/2024] [Indexed: 06/19/2024] Open
Abstract
The 16-subunit Constitutive Centromere-associated Network (CCAN)-based inner kinetochore is well-known for connecting centromeric chromatin to the spindle-binding outer kinetochore. Here, we report a non-canonical role for the inner kinetochore in directly regulating sister-chromatid cohesion at centromeres. We provide biochemical, X-ray crystal structure, and intracellular ectopic localization evidence that the inner kinetochore directly binds cohesin, a ring-shaped multi-subunit complex that holds sister chromatids together from S-phase until anaphase onset. This interaction is mediated by binding of the 5-subunit CENP-OPQUR sub-complex of CCAN to the Scc1-SA2 sub-complex of cohesin. Mutation in the CENP-U subunit of the CENP-OPQUR complex that abolishes its binding to the composite interface between Scc1 and SA2 weakens centromeric cohesion, leading to premature separation of sister chromatids during delayed metaphase. We further show that CENP-U competes with the cohesin release factor Wapl for binding the interface of Scc1-SA2, and that the cohesion-protecting role for CENP-U can be bypassed by depleting Wapl. Taken together, this study reveals an inner kinetochore-bound pool of cohesin, which strengthens centromeric sister-chromatid cohesion to resist metaphase spindle pulling forces.
Collapse
Affiliation(s)
- Lu Yan
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
| | - Xueying Yuan
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
| | - Mingjie Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qinfu Chen
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Miao Zhang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
| | - Junfen Xu
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Long Zhang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Jun Huang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Weiguo Lu
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Xiaojing He
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Haiyan Yan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Fangwei Wang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China.
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
- Zhejiang Provincial Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
28
|
Li P, Li Y, Ma X, Li L, Zeng S, Peng Y, Liang H, Zhang G. Identification of naphthalimide-derivatives as novel PBD-targeted polo-like kinase 1 inhibitors with efficacy in drug-resistant lung cancer cells. Eur J Med Chem 2024; 271:116416. [PMID: 38657480 DOI: 10.1016/j.ejmech.2024.116416] [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: 02/01/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
Targeting polo-box domain (PBD) small molecule for polo-like kinase 1 (PLK1) inhibition is a viable alternative to target kinase domain (KD), which could avoid pan-selectivity and dose-limiting toxicity of ATP-competitive inhibitors. However, their efficacy in these settings is still low and inaccessible to clinical requirement. Herein, we utilized a structure-based high-throughput virtual screen to find novel chemical scaffold capable of inhibiting PLK1 via targeting PBD and identified an initial hit molecule compound 1a. Based on the lead compound 1a, a structural optimization approach was carried out and several series of derivatives with naphthalimide structural motif were synthesized. Compound 4Bb was identified as a new potent PLK1 inhibitor with a KD value of 0.29 μM. 4Bb could target PLK1 PBD to inhibit PLK1 activity and subsequently suppress the interaction of PLK1 with protein regulator of cytokinesis 1 (PRC1), finally leading to mitotic catastrophe in drug-resistant lung cancer cells. Furthermore, 4Bb could undergo nucleophilic substitution with the thiol group of glutathione (GSH) to disturb the redox homeostasis through exhausting GSH. By regulating cell cycle machinery and increasing cellular oxidative stress, 4Bb exhibited potent cytotoxicity to multiple cancer cells and drug-resistant cancer cells. Subcutaneous and oral administration of 4Bb could effectively inhibit the growth of drug-resistant tumors in vivo, doubling the survival time of tumor bearing mice without side effects in normal tissues. Thus, our study offers an orally-available, structurally-novel PLK1 inhibitor for drug-resistant lung cancer therapy.
Collapse
Affiliation(s)
- Pingping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Yongkun Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Xuesong Ma
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Liangping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Shulan Zeng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Yan Peng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China.
| | - Guohai Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China.
| |
Collapse
|
29
|
Black EM, Ramírez Parrado CA, Trier I, Li W, Joo YK, Pichurin J, Liu Y, Kabeche L. Chk2 sustains PLK1 activity in mitosis to ensure proper chromosome segregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584115. [PMID: 38559033 PMCID: PMC10979866 DOI: 10.1101/2024.03.08.584115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Polo-like kinase 1 (PLK1) protects against genome instability by ensuring timely and accurate mitotic cell division. PLK1 activity is tightly regulated throughout the cell cycle. Although the pathways that initially activate PLK1 in G2 are well-characterized, the factors that directly regulate PLK1 in mitosis remain poorly understood. Here, we identify that human PLK1 activity is sustained by the DNA damage response kinase Checkpoint kinase 2 (Chk2) in mitosis. Chk2 directly phosphorylates PLK1 T210, a residue on its T-loop whose phosphorylation is essential for full PLK1 kinase activity. Loss of Chk2-dependent PLK1 activity causes increased mitotic errors, including chromosome misalignment, chromosome missegregation, and cytokinetic defects. Moreover, Chk2 deficiency increases sensitivity to PLK1 inhibitors, suggesting that Chk2 status may be an informative biomarker for PLK1 inhibitor efficacy. This work demonstrates that Chk2 sustains mitotic PLK1 activity and protects genome stability through discrete functions in interphase DNA damage repair and mitotic chromosome segregation.
Collapse
|
30
|
Nelson CB, Rogers S, Roychoudhury K, Tan YS, Atkinson CJ, Sobinoff AP, Tomlinson CG, Hsu A, Lu R, Dray E, Haber M, Fletcher JI, Cesare AJ, Hegde RS, Pickett HA. The Eyes Absent family members EYA4 and EYA1 promote PLK1 activation and successful mitosis through tyrosine dephosphorylation. Nat Commun 2024; 15:1385. [PMID: 38360978 PMCID: PMC10869800 DOI: 10.1038/s41467-024-45683-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/31/2024] [Indexed: 02/17/2024] Open
Abstract
The Eyes Absent proteins (EYA1-4) are a biochemically unique group of tyrosine phosphatases known to be tumour-promoting across a range of cancer types. To date, the targets of EYA phosphatase activity remain largely uncharacterised. Here, we identify Polo-like kinase 1 (PLK1) as an interactor and phosphatase substrate of EYA4 and EYA1, with pY445 on PLK1 being the primary target site. Dephosphorylation of pY445 in the G2 phase of the cell cycle is required for centrosome maturation, PLK1 localization to centrosomes, and polo-box domain (PBD) dependent interactions between PLK1 and PLK1-activation complexes. Molecular dynamics simulations support the rationale that pY445 confers a structural impairment to PBD-substrate interactions that is relieved by EYA-mediated dephosphorylation. Depletion of EYA4 or EYA1, or chemical inhibition of EYA phosphatase activity, dramatically reduces PLK1 activation, causing mitotic defects and cell death. Overall, we have characterized a phosphotyrosine signalling network governing PLK1 and mitosis.
Collapse
Affiliation(s)
- Christopher B Nelson
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Samuel Rogers
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Kaushik Roychoudhury
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Caroline J Atkinson
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Alexander P Sobinoff
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Christopher G Tomlinson
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Anton Hsu
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Robert Lu
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Eloise Dray
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Anthony J Cesare
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Rashmi S Hegde
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hilda A Pickett
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
31
|
Velez-Aguilera G, Ossareh-Nazari B, Pintard L. Dissecting the Multiple Functions of the Polo-Like Kinase 1 in the C. elegans Zygote. Methods Mol Biol 2024; 2740:63-88. [PMID: 38393469 DOI: 10.1007/978-1-0716-3557-5_4] [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] [Indexed: 02/25/2024]
Abstract
Plk1 (polo-like kinase 1) is an evolutionarily conserved serine/threonine kinase instrumental for mitotic entry and progression. Beyond these canonical functions, Plk1 also regulates cell polarization and cell fate during asymmetric cell divisions in C. elegans and D. melanogaster. Plk1 contains a specialized phosphoserine-threonine binding domain, the polo-box domain (PBD), which localizes and concentrates the kinase at its various sites of action within the cell in space and time. Here we present protocols to express and purify the C. elegans Plk1 kinase along with biochemical and phosphoproteomic approaches to interrogate the PBD interactome and to dissect Plk1 substrate interactions. These protocols are most suitable for the identification of Plk1 targets in C. elegans embryos but can be easily adapted to identify and study Plk1 substrates from any source."
Collapse
Affiliation(s)
- Griselda Velez-Aguilera
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- Programme Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Batool Ossareh-Nazari
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- Programme Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Lionel Pintard
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France.
- Programme Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
32
|
Gao J, Huang W, Zhao S, Wang R, Wang Z, Ye J, Lin L, Cai W, Mi Y. Polo-like kinase 1 inhibitor NMS-P937 represses nasopharyngeal carcinoma progression via induction of mitotic abnormalities. J Biochem Mol Toxicol 2024; 38:e23590. [PMID: 38037286 DOI: 10.1002/jbt.23590] [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: 07/19/2022] [Revised: 02/05/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
Polo-like kinase 1 (PLK1) inhibitor NMS-P937 is a targeted therapeutic agent with good preclinical efficacy in various human cancers, and its therapeutic effect on nasopharyngeal carcinoma (NPC) remains to be determined. Here, to explore biological activity of NMS-P937 in NPC, multiple types of NPC cells were utilized. We tested IC50 values, carried out flow cytometry, western blot analysis analysis, immunofluorescence, and constructed subcutaneous xenograft mouse models. We found that treatment with NMS-P937 increased the proportion of G2/M phase NPC cells, where CyclinB1 expression was upregulated and CyclinE1 expression was downregulated. Besides, NMS-P937 treatment-induced NPC cell apoptosis with increased cleavage of PARP and caspase-3. Mechanistically, NMS-P937 treatment led to aberrant mitosis, causing increased reactive oxygen species (ROS) levels. ROS scavenger N-acetylcysteine partially reversed ROS levels induced by NMS-P937. Furthermore, NMS-P937 administration restrained NPC xenografts growth in nude mice. Overall, NMS-P937 suppressed NPC cell proliferation and increased ROS levels, causing cell cycle abnormalities and apoptosis. NMS-P937 holds great promise as a therapeutic agent for treating nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Jing Gao
- Department of Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Fujian, China
| | - Weirong Huang
- Department of Medical Oncology, Xiamen Key Laboratory of Thoracic Tumor Diagnosis and Treatment, School of Clinical Medicine, Institute of Lung Cancer, The First Affiliated Hospital of Xiamen University, Fujian Medical University, Fujian, China
| | - Senxia Zhao
- Department of Medical Oncology, Xiamen Key Laboratory of Thoracic Tumor Diagnosis and Treatment, School of Clinical Medicine, Institute of Lung Cancer, The First Affiliated Hospital of Xiamen University, Fujian Medical University, Fujian, China
| | - Rong Wang
- School of Medicine, Guangxi University, Nanning, China
| | - Zhilin Wang
- Department of Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Fujian, China
| | - Juanping Ye
- Department of Medical Oncology, Xiamen Key Laboratory of Thoracic Tumor Diagnosis and Treatment, School of Clinical Medicine, Institute of Lung Cancer, The First Affiliated Hospital of Xiamen University, Fujian Medical University, Fujian, China
| | - Lie Lin
- Department of Radiotherapy, The First Affiliated Hospital of Xiamen University, Fujian, China
| | - Weifeng Cai
- Department of Medical Oncology, Xiamen Key Laboratory of Thoracic Tumor Diagnosis and Treatment, School of Clinical Medicine, Institute of Lung Cancer, The First Affiliated Hospital of Xiamen University, Fujian Medical University, Fujian, China
| | - Yanjun Mi
- Department of Medical Oncology, Xiamen Key Laboratory of Thoracic Tumor Diagnosis and Treatment, School of Clinical Medicine, Institute of Lung Cancer, The First Affiliated Hospital of Xiamen University, Fujian Medical University, Fujian, China
| |
Collapse
|
33
|
Nair VM, Sabu AS, Hussain A, Kombarakkaran DP, Lakshmi RB, Manna TK. E3-ubiquitin ligase, FBXW7 regulates mitotic progression by targeting BubR1 for ubiquitin-mediated degradation. Cell Mol Life Sci 2023; 80:374. [PMID: 38008853 PMCID: PMC11072012 DOI: 10.1007/s00018-023-05019-9] [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: 07/09/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/28/2023]
Abstract
Faithful chromosome segregation requires correct attachment of kinetochores with the spindle microtubules. Erroneously-attached kinetochores recruit proteins to activate Spindle assembly checkpoint (SAC), which senses the errors and signals cells to delay anaphase progression for error correction. Temporal control of the levels of SAC activating-proteins is critical for checkpoint activation and silencing, but its mechanism is not fully understood. Here, we show that E3 ubiquitin ligase, SCF-FBXW7 targets BubR1 for ubiquitin-mediated degradation and thereby controls SAC in human cells. Depletion of FBXW7 results in prolonged metaphase arrest with increased stabilization of BubR1 at kinetochores. Similar kinetochore stabilization is also observed for BubR1-interacting protein, CENP-E. FBXW7 induced ubiquitination of both BubR1 and the BubR1-interacting kinetochore-targeting domain of CENP-E, but CENP-E domain degradation is dependent on BubR1. Interestingly, Cdk1 inhibition disrupts FBXW7-mediated BubR1 targeting and further, phospho-resistant mutation of Cdk1-targeted phosphorylation site, Thr 620 impairs BubR1-FBXW7 interaction and FBXW7-mediated BubR1 ubiquitination, supporting its role as a phosphodegron for FBXW7. The results demonstrate SCF-FBXW7 as a key regulator of spindle assembly checkpoint that controls stability of BubR1 and its associated CENP-E at kinetochores. They also support that upstream Cdk1 specific BubR1 phosphorylation signals the ligase to activate the process.
Collapse
Affiliation(s)
- Vishnu M Nair
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram, Kerala, 695551, India
| | - Amit Santhu Sabu
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram, Kerala, 695551, India
| | - Ahmed Hussain
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram, Kerala, 695551, India
| | - Delvin P Kombarakkaran
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram, Kerala, 695551, India
| | - R Bhagya Lakshmi
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram, Kerala, 695551, India
| | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram, Kerala, 695551, India.
| |
Collapse
|
34
|
Wang PC, Yang ZS, Gu XW. Effect of Aurora kinase B on polyploidy and decidualization in mouse uterus. Am J Reprod Immunol 2023; 90:e13793. [PMID: 37881124 DOI: 10.1111/aji.13793] [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: 06/19/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023] Open
Abstract
RESEARCH QUESTION Decidualization is critical to the establishment of mouse normal pregnancy. The fibroblast-like stromal cells in the process form polyploid multinucleated cells. Aurora kinase B (Aurora B) has previously been shown to regulate polyploidy in various cells. However, whether Aurora B regulates the formation of decidual cell polyploidization and its regulatory mechanisms remain poorly understood. DESIGN Establish decidualization model of mouse primary endometrial stromal cells in vitro. Construct pseudopregnancy mouse models and delayed-activation mouse models. Detect Aurora B and polyploidization related genes in mouse uteri treated by Aurora B specific inhibitor Barasertib and CPT. RESULTS In this study, we found that Aurora B was strongly expressed in endometrial stromal cells after implantation. Additionally, Aurora B was remarkably up regulated in the stromal cells of oil-induced deciduomoa and in vitro decidualization. As an Aurora B specific inhibitor, Barasertib significantly inhibits the mRNA expression of Prl8a2, a marker of mouse decidualization. Furthermore, the protein levels of p-Plk1, Survivin and p-Cdk1 were inhibited by Barasertib. CPT-induced DNA damage suppressed Aurkb (encodes Aurora B) expression, thus resulting in polyploidization. CONCLUSION Our data shows that Aurora B is expressed in decidual stromal cells of implantation sites and plays a key role for mouse decidualization. The protein of Plk1, Survivn, and Cdk1 may participate in formation of decidual cell polyploidization during mouse decidualization.
Collapse
Affiliation(s)
- Peng-Chao Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Zhen-Shan Yang
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Xiao-Wei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| |
Collapse
|
35
|
Jema S, Chen C, Humphrey L, Karmarkar S, Ferrari F, Joglekar AP. Signaling protein abundance modulates the strength of the spindle assembly checkpoint. Curr Biol 2023; 33:4505-4515.e4. [PMID: 37738972 PMCID: PMC10615864 DOI: 10.1016/j.cub.2023.08.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/19/2023] [Accepted: 08/24/2023] [Indexed: 09/24/2023]
Abstract
During mitosis, unattached kinetochores in a dividing cell signal to the spindle assembly checkpoint (SAC) to delay anaphase onset and prevent chromosome missegregation.1,2,3,4 The signaling activity of these kinetochores and the likelihood of chromosome missegregation depend on the amount of SAC signaling proteins each kinetochore recruits.5,6,7,8 Therefore, factors that control SAC protein recruitment must be thoroughly understood. Phosphoregulation of kinetochore and SAC signaling proteins due to the concerted action of many kinases and phosphatases is a significant determinant of the SAC protein recruitment to signaling kinetochores.9 Whether the abundance of SAC proteins also influences the recruitment and signaling activity of human kinetochores has not been studied.8,10 Here, we reveal that the low cellular abundance of the SAC signaling protein Bub1 limits its own recruitment and that of BubR1 and restricts the SAC signaling activity of the kinetochore. Conversely, Bub1 overexpression results in higher recruitment of SAC proteins, producing longer delays in anaphase onset. We also find that the number of SAC proteins recruited by a signaling kinetochore is inversely correlated with the total number of signaling kinetochores in the cell. This correlation likely arises from the competition among the signaling kinetochores to recruit from a limited pool of signaling proteins, including Bub1. The inverse correlation may allow the dividing cell to prevent a large number of signaling kinetochores in early prophase from generating an overly large signal while enabling the last unaligned kinetochore in late prometaphase to signal at the maximum strength.
Collapse
Affiliation(s)
- Soubhagyalaxmi Jema
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Chu Chen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lauren Humphrey
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Shriya Karmarkar
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Frank Ferrari
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ajit P Joglekar
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
36
|
Wang Z, Xia Y, Wang Y, Zhu R, Li H, Liu Y, Shen N. The E3 ligase TRIM26 suppresses ferroptosis through catalyzing K63-linked ubiquitination of GPX4 in glioma. Cell Death Dis 2023; 14:695. [PMID: 37872147 PMCID: PMC10593845 DOI: 10.1038/s41419-023-06222-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
The selenium-containing enzyme GPX4 moonlights as a central regulator of ferroptosis, an iron-dependent, nonapoptotic form of regulated cell death caused by lipid peroxidation. Yet, little is known about the mechanisms underlying the regulation of its post-transcriptional modifications. Here, we identify the tripartite motif-containing protein TRIM26 as an E3 ubiquitin ligase of GPX4. TRIM26 directly interacts with GPX4 through its Ring domain and catalyzes the ubiquitination of GPX4 at K107 and K117, which promotes the switch in polyubiquitination of GPX4 from K48 to K63, thus enhancing GPX4 protein stability. Moreover, PLK1-mediated S127 phosphorylation of TRIM26 enhances the interaction between TRIM26 and GPX4. Inhibition of TRIM26 phosphorylation causes a reduction in GPX4 K63-linked polyubiquitination and diminishes GPX4 protein levels in tumor cells. Further investigation revealed that TRIM26 is overexpressed in glioma cells. TRIM26 silencing dramatically impedes ferroptosis resistance and tumorigenesis in glioma in vivo and in vitro. Clinically, TRIM26 expression shows a direct correlation with GPX4 and PLK1 levels in glioma samples and is associated with poor outcome in patients with glioma. Collectively, these findings define the role of GPX4 K63-linked polyubiquitination in ferroptosis and suggest a potential strategy for glioma treatment.
Collapse
Affiliation(s)
- Zhangjie Wang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuan Xia
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Yang Wang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ruiqiu Zhu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Hongbo Li
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Yu Liu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Na Shen
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
37
|
Corno A, Cordeiro MH, Allan LA, Lim Q, Harrington E, Smith RJ, Saurin AT. A bifunctional kinase-phosphatase module balances mitotic checkpoint strength and kinetochore-microtubule attachment stability. EMBO J 2023; 42:e112630. [PMID: 37712330 PMCID: PMC10577578 DOI: 10.15252/embj.2022112630] [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: 09/16/2022] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
Two major mechanisms safeguard genome stability during mitosis: the mitotic checkpoint delays mitosis until all chromosomes have attached to microtubules, and the kinetochore-microtubule error-correction pathway keeps this attachment process free from errors. We demonstrate here that the optimal strength and dynamics of these processes are set by a kinase-phosphatase pair (PLK1-PP2A) that engage in negative feedback from adjacent phospho-binding motifs on the BUB complex. Uncoupling this feedback to skew the balance towards PLK1 produces a strong checkpoint, hypostable microtubule attachments and mitotic delays. Conversely, skewing the balance towards PP2A causes a weak checkpoint, hyperstable microtubule attachments and chromosome segregation errors. These phenotypes are associated with altered BUB complex recruitment to KNL1-MELT motifs, implicating PLK1-PP2A in controlling auto-amplification of MELT phosphorylation. In support, KNL1-BUB disassembly becomes contingent on PLK1 inhibition when KNL1 is engineered to contain excess MELT motifs. This elevates BUB-PLK1/PP2A complex levels on metaphase kinetochores, stabilises kinetochore-microtubule attachments, induces chromosome segregation defects and prevents KNL1-BUB disassembly at anaphase. Together, these data demonstrate how a bifunctional PLK1/PP2A module has evolved together with the MELT motifs to optimise BUB complex dynamics and ensure accurate chromosome segregation.
Collapse
Affiliation(s)
- Andrea Corno
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Marilia H Cordeiro
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Lindsey A Allan
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Qian‐Wei Lim
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Elena Harrington
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Richard J Smith
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Adrian T Saurin
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| |
Collapse
|
38
|
Zhao S, Li Y, Li G, Ye J, Wang R, Zhang X, Li F, Gao C, Li J, Jiang J, Mi Y. PI3K/mTOR inhibitor VS-5584 combined with PLK1 inhibitor exhibits synergistic anti-cancer effects on non-small cell lung cancer. Eur J Pharmacol 2023; 957:176004. [PMID: 37625683 DOI: 10.1016/j.ejphar.2023.176004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/30/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Small molecule drugs are of significant importance in the treatment of non-small cell lung cancer (NSCLC). Here, we explored biological effects of the PI3K/mTOR inhibitor VS-5584 on NSCLC. Our findings indicated that VS-5584 administration resulted in a dose-dependent inhibition of NSCLC cell proliferation, as well as the induction of apoptosis and cycle arrest. Additionally, we observed a significant increase in intracellular reactive oxygen species (ROS) levels following VS-5584 treatment. The use of the ROS inhibitor N-acetylcysteine (NAC) effectively reduced ROS levels and decreased the proportion of apoptotic cells. Treatment with VS-5584 led to an upregulation of genes associated with apoptosis and cell cycle, such as c-caspase 3 and P21. Conversely, a downregulation of cyclin-dependent kinase 1 (CDK1) expression was observed. Next, transcriptome analyses revealed that VS-5584 treatment altered the abundance of 1520 genes/transcripts in PC-9 cells, one of which was polo-like kinase 1 (PLK1). These differentially expressed genes were primarily enriched in biological processes such as cell cycle regulation and cell apoptosis, which are closely linked to the P53 and apoptosis pathways. Co-treatment with VS-5584 and PLK1 inhibitor NMS-P937 resulted in enhanced cancer cell death, exhibiting synergistic inhibitory activity. Notably, VS-5584 inhibited the growth of NSCLC in a patient-derived xenograft (PDX) mouse model without observable abnormalities in major organs. Overall, VS-5584 effectively suppressed the growth of NSCLC cells both in vitro and in vivo. VS-5584 combined with NMS-P937 exhibited a synergistic effect in inhibiting NSCLC cell growth. These findings suggest that VS-5584 has potential as a therapeutic strategy for treating NSCLC.
Collapse
Affiliation(s)
- Senxia Zhao
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Yibin Li
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Gang Li
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Juanping Ye
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Rong Wang
- Department of Thoracic Surgery, Xiamen Key Laboratory of Thoracic Tumor Diagnosis and Treatment, Institute of Lung Cancer, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Xiaoting Zhang
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Fei Li
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Chang Gao
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Junbiao Li
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China
| | - Jie Jiang
- Department of Thoracic Surgery, Xiamen Key Laboratory of Thoracic Tumor Diagnosis and Treatment, Institute of Lung Cancer, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China.
| | - Yanjun Mi
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Clinical Medicine, Xiamen University, Xiamen, 361003, Fujian Province, PR China.
| |
Collapse
|
39
|
Dudka D, Akins RB, Lampson MA. FREEDA: An automated computational pipeline guides experimental testing of protein innovation. J Cell Biol 2023; 222:e202212084. [PMID: 37358475 PMCID: PMC10292211 DOI: 10.1083/jcb.202212084] [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: 12/17/2022] [Revised: 04/22/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023] Open
Abstract
Cell biologists typically focus on conserved regions of a protein, overlooking innovations that can shape its function over evolutionary time. Computational analyses can reveal potential innovations by detecting statistical signatures of positive selection that lead to rapid accumulation of beneficial mutations. However, these approaches are not easily accessible to non-specialists, limiting their use in cell biology. Here, we present an automated computational pipeline FREEDA that provides a simple graphical user interface requiring only a gene name; integrates widely used molecular evolution tools to detect positive selection in rodents, primates, carnivores, birds, and flies; and maps results onto protein structures predicted by AlphaFold. Applying FREEDA to >100 centromere proteins, we find statistical evidence of positive selection within loops and turns of ancient domains, suggesting innovation of essential functions. As a proof-of-principle experiment, we show innovation in centromere binding of mouse CENP-O. Overall, we provide an accessible computational tool to guide cell biology research and apply it to experimentally demonstrate functional innovation.
Collapse
Affiliation(s)
- Damian Dudka
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - R. Brian Akins
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A. Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Brambati A, Sacco O, Porcella S, Heyza J, Kareh M, Schmidt JC, Sfeir A. RHINO directs MMEJ to repair DNA breaks in mitosis. Science 2023; 381:653-660. [PMID: 37440612 PMCID: PMC10561558 DOI: 10.1126/science.adh3694] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
Nonhomologous end-joining (NHEJ) and homologous recombination (HR) are the primary pathways for repairing DNA double-strand breaks (DSBs) during interphase, whereas microhomology-mediated end-joining (MMEJ) has been regarded as a backup mechanism. Through CRISPR-Cas9-based synthetic lethal screens in cancer cells, we identified subunits of the 9-1-1 complex (RAD9A-RAD1-HUS1) and its interacting partner, RHINO, as crucial MMEJ factors. We uncovered an unexpected function for RHINO in restricting MMEJ to mitosis. RHINO accumulates in M phase, undergoes Polo-like kinase 1 (PLK1) phosphorylation, and interacts with polymerase θ (Polθ), enabling its recruitment to DSBs for subsequent repair. Additionally, we provide evidence that MMEJ activity in mitosis repairs persistent DSBs that originate in S phase. Our findings offer insights into the synthetic lethal relationship between the genes POLQ and BRCA1 and BRAC2 and the synergistic effect of Polθ and poly(ADP-ribose) polymerase (PARP) inhibitors.
Collapse
Affiliation(s)
- Alessandra Brambati
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Olivia Sacco
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Sarina Porcella
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Joshua Heyza
- Institute for Quantitative Health Sciences and Engineering, Michigan State University; East Lansing, MI, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University; East Lansing, MI, USA
| | - Mike Kareh
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Jens C. Schmidt
- Institute for Quantitative Health Sciences and Engineering, Michigan State University; East Lansing, MI, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University; East Lansing, MI, USA
| | - Agnel Sfeir
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| |
Collapse
|
41
|
Yatskevich S, Barford D, Muir KW. Conserved and divergent mechanisms of inner kinetochore assembly onto centromeric chromatin. Curr Opin Struct Biol 2023; 81:102638. [PMID: 37343495 DOI: 10.1016/j.sbi.2023.102638] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023]
Abstract
Kinetochores are large protein complexes built on centromeric chromatin that mediate chromosome segregation. The inner kinetochore, or constitutive centromere-associated network (CCAN), assembles onto centromeres defined by centromere protein A (CENP-A) nucleosomes (CENP-ANuc), and acts as a platform for the regulated assembly of the microtubule-binding outer kinetochore. Recent cryo-EM work revealed structural conservation of CCAN, from the repeating human regional centromeres to the point centromere of budding yeast. Centromere recognition is determined mainly through engagement of duplex DNA proximal to the CENP-A nucleosome by a DNA-binding CENP-LN channel located at the core of CCAN. Additional DNA interactions formed by other CCAN modules create an enclosed DNA-binding chamber. This configuration explains how kinetochores maintain their tight grip on centromeric DNA to withstand the forces of chromosome segregation. Defining the higher-order architecture of complete kinetochore assemblies with implications for understanding the 3D organisation of regional centromeres and mechanisms of kinetochore dynamics, including how kinetochores sense and respond to tension, are important future directions.
Collapse
Affiliation(s)
- Stanislau Yatskevich
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom. https://twitter.com/StanislauY
| | - David Barford
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom.
| | - Kyle W Muir
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom. https://twitter.com/centromuir
| |
Collapse
|
42
|
Normandin K, Coulombe-Huntington J, St-Denis C, Bernard A, Bourouh M, Bertomeu T, Tyers M, Archambault V. Genetic enhancers of partial PLK1 inhibition reveal hypersensitivity to kinetochore perturbations. PLoS Genet 2023; 19:e1010903. [PMID: 37639469 PMCID: PMC10491399 DOI: 10.1371/journal.pgen.1010903] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/08/2023] [Accepted: 08/06/2023] [Indexed: 08/31/2023] Open
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase required for mitosis and cytokinesis. As cancer cells are often hypersensitive to partial PLK1 inactivation, chemical inhibitors of PLK1 have been developed and tested in clinical trials. However, these small molecule inhibitors alone are not completely effective. PLK1 promotes numerous molecular and cellular events in the cell division cycle and it is unclear which of these events most crucially depend on PLK1 activity. We used a CRISPR-based genome-wide screening strategy to identify genes whose inactivation enhances cell proliferation defects upon partial chemical inhibition of PLK1. Genes identified encode proteins that are functionally linked to PLK1 in multiple ways, most notably factors that promote centromere and kinetochore function. Loss of the kinesin KIF18A or the outer kinetochore protein SKA1 in PLK1-compromised cells resulted in mitotic defects, activation of the spindle assembly checkpoint and nuclear reassembly defects. We also show that PLK1-dependent CENP-A loading at centromeres is extremely sensitive to partial PLK1 inhibition. Our results suggest that partial inhibition of PLK1 compromises the integrity and function of the centromere/kinetochore complex, rendering cells hypersensitive to different kinetochore perturbations. We propose that KIF18A is a promising target for combinatorial therapies with PLK1 inhibitors.
Collapse
Affiliation(s)
- Karine Normandin
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | | | - Corinne St-Denis
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Alexandre Bernard
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Mohammed Bourouh
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Thierry Bertomeu
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Département de médecine, Université de Montréal, Montréal, Canada
| | - Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Canada
| |
Collapse
|
43
|
Chapagai D, Merhej G, McInnes C, Wyatt MD. Structural Basis for Variations in Polo-like Kinase 1 Conformation and Intracellular Stability Induced by ATP-Competitive and Novel Noncompetitive Abbapolin Inhibitors. ACS Chem Biol 2023; 18:1642-1652. [PMID: 37433100 PMCID: PMC11295584 DOI: 10.1021/acschembio.3c00269] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Polo-like kinase 1 (PLK1) is an essential protein kinase with multiple roles in mitotic progression. PLK1 consists of a kinase domain (KD) and a phosphopeptide-binding polobox domain (PBD), which is responsible for substrate recognition and subcellular localization. The regulation of PLK1 involves an autoinhibitory conformation in which KD and PBD interact. Our previous work identified PBD-binding molecules termed abbapolins that inhibit the cellular phosphorylation of a PLK1 substrate and induce the loss of intracellular PLK1. Here, we describe a comparison of the abbapolin activity with that of KD inhibitors to gain insight into conformational features of PLK1. As measured by a cellular thermal shift assay, abbapolins produce ligand-induced thermal stabilization of PLK1. In contrast, KD inhibitors decreased the soluble PLK1, suggesting that catalytic-site binding causes a less thermally stable PLK1 conformation. Binding measurements with full-length PLK1 and a KD inhibitor also demonstrated a conformational change. Interestingly, the cellular consequences of KD versus PBD engagement contrast as KD binding causes the accumulation of intracellular PLK1, whereas PBD binding produces a striking loss of nuclear PLK1. These data are consistent with the relief of autoinhibited PLK1 by KD binders; an explanation for these observations is presented using structures for the catalytic domain and full-length PLK1 predicted by AlphaFold. Collectively, the results highlight an underappreciated aspect of targeting PLK1, namely, conformational perturbations induced by KD versus PBD binding. In addition to their significance for PBD-binding ligands, these observations have implications for the development of ATP-competitive PLK1 inhibitors because catalytic inhibitors may conversely promote PLK1 noncatalytic functions, which may explain their lack of clinical efficacy to date.
Collapse
Affiliation(s)
| | - George Merhej
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208
| | - Campbell McInnes
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208
| | - Michael D. Wyatt
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208
| |
Collapse
|
44
|
Nkombo Nkoula S, Velez-Aguilera G, Ossareh-Nazari B, Van Hove L, Ayuso C, Legros V, Chevreux G, Thomas L, Seydoux G, Askjaer P, Pintard L. Mechanisms of nuclear pore complex disassembly by the mitotic Polo-like kinase 1 (PLK-1) in C. elegans embryos. SCIENCE ADVANCES 2023; 9:eadf7826. [PMID: 37467327 PMCID: PMC10355831 DOI: 10.1126/sciadv.adf7826] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023]
Abstract
The nuclear envelope, which protects and organizes the genome, is dismantled during mitosis. In the Caenorhabditis elegans zygote, nuclear envelope breakdown (NEBD) of the parental pronuclei is spatially and temporally regulated during mitosis to promote the unification of the maternal and paternal genomes. Nuclear pore complex (NPC) disassembly is a decisive step of NEBD, essential for nuclear permeabilization. By combining live imaging, biochemistry, and phosphoproteomics, we show that NPC disassembly is a stepwise process that involves Polo-like kinase 1 (PLK-1)-dependent and -independent steps. PLK-1 targets multiple NPC subcomplexes, including the cytoplasmic filaments, central channel, and inner ring. PLK-1 is recruited to and phosphorylates intrinsically disordered regions (IDRs) of several multivalent linker nucleoporins. Notably, although the phosphosites are not conserved between human and C. elegans nucleoporins, they are located in IDRs in both species. Our results suggest that targeting IDRs of multivalent linker nucleoporins is an evolutionarily conserved driver of NPC disassembly during mitosis.
Collapse
Affiliation(s)
- Sylvia Nkombo Nkoula
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Programme Équipe Labellisée Ligue contre le Cancer, Paris, France
| | - Griselda Velez-Aguilera
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Programme Équipe Labellisée Ligue contre le Cancer, Paris, France
| | - Batool Ossareh-Nazari
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Programme Équipe Labellisée Ligue contre le Cancer, Paris, France
| | - Lucie Van Hove
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Programme Équipe Labellisée Ligue contre le Cancer, Paris, France
| | - Cristina Ayuso
- Andalusian Center for Developmental Biology (CABD), CSIC/JA/Universidad Pablo de Olavide, Seville, Spain
| | - Véronique Legros
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Guillaume Chevreux
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Laura Thomas
- HHMI and Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Géraldine Seydoux
- HHMI and Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter Askjaer
- Andalusian Center for Developmental Biology (CABD), CSIC/JA/Universidad Pablo de Olavide, Seville, Spain
| | - Lionel Pintard
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Programme Équipe Labellisée Ligue contre le Cancer, Paris, France
| |
Collapse
|
45
|
Tian Y, Yu B, Zhang Y, Zhang S, Lv B, Gong S, Li J. Exploration of the potential common pathogenic mechanisms in COVID-19 and silicosis by using bioinformatics and system biology. Funct Integr Genomics 2023; 23:199. [PMID: 37278873 PMCID: PMC10241611 DOI: 10.1007/s10142-023-01092-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023]
Abstract
Silicosis is an occupational lung disease that is common worldwide. In recent years, coronavirus disease 2019 (COVID-19) has provided daunting challenges to public healthcare systems globally. Although multiple studies have shown a close link between COVID-19 and other respiratory diseases, the inter-relational mechanisms between COVID-19 and silicosis remain unclear. This study aimed to explore the shared molecular mechanisms and drug targets of COVID-19 and silicosis. Gene expression profiling identified four modules that were most closely associated with both diseases. Furthermore, we performed functional analysis and constructed a protein-protein interaction network. Seven hub genes (budding uninhibited by benzimidazoles 1 [BUB1], protein regulator of cytokinesis 1 [PRC1], kinesin family member C1 [KIFC1], ribonucleotide reductase regulatory subunit M2 [RRM2], cyclin-dependent kinase inhibitor 3 [CDKN3], Cyclin B2 [CCNB2], and minichromosome maintenance complex component 6 [MCM6]) were involved in the interaction between COVID-19 and silicosis. We investigated how diverse microRNAs and transcription factors regulate these seven genes. Subsequently, the correlation between the hub genes and infiltrating immune cells was explored. Further in-depth analyses were performed based on single-cell transcriptomic data from COVID-19, and the expression of hub-shared genes was characterized and located in multiple cell clusters. Finally, molecular docking results reveal small molecular compounds that may improve COVID-19 and silicosis. The current study reveals the common pathogenesis of COVID-19 and silicosis, which may provide a novel reference for further research.
Collapse
Affiliation(s)
- Yunze Tian
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, Xi'an, 710004, China
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, Xi'an, 710004, China
| | - Beibei Yu
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, Xi'an, 710004, China
| | - Yongfeng Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, Xi'an, 710004, China
| | - Sanpeng Zhang
- Operating room, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, 710004, Xi'an, China
| | - Boqiang Lv
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, Xi'an, 710004, China
| | - Shouping Gong
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, Xi'an, 710004, China.
| | - Jianzhong Li
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiao Tong University, Shaanxi Province, Xi'an, 710004, China.
| |
Collapse
|
46
|
Davies T, Hardwick KG. Cell-cycle control: Timing is everything for the Plk1-Bub1 partnership. Curr Biol 2023; 33:R447-R449. [PMID: 37279667 DOI: 10.1016/j.cub.2023.04.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Bub1 and Polo kinases are well-known multitasking regulators of mitosis. New work shows that they team up at kinetochores to determine the mitotic duration of embryonic divisions in nematodes. As is often the case, the key effector is Cdc20 activity.
Collapse
Affiliation(s)
- Thomas Davies
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Kevin G Hardwick
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, UK.
| |
Collapse
|
47
|
Houston J, Ohta M, Gómez-Cavazos JS, Deep A, Corbett KD, Oegema K, Lara-Gonzalez P, Kim T, Desai A. BUB-1-bound PLK-1 directs CDC-20 kinetochore recruitment to ensure timely embryonic mitoses. Curr Biol 2023; 33:2291-2299.e10. [PMID: 37137308 PMCID: PMC10270731 DOI: 10.1016/j.cub.2023.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/13/2023] [Accepted: 04/12/2023] [Indexed: 05/05/2023]
Abstract
During mitosis, chromosomes assemble kinetochores to dynamically couple with spindle microtubules.1,2 Kinetochores also function as signaling hubs directing mitotic progression by recruiting and controlling the fate of the anaphase promoting complex/cyclosome (APC/C) activator CDC-20.3,4,5 Kinetochores either incorporate CDC-20 into checkpoint complexes that inhibit the APC/C or dephosphorylate CDC-20, which allows it to interact with and activate the APC/C.4,6 The importance of these two CDC-20 fates likely depends on the biological context. In human somatic cells, the major mechanism controlling mitotic progression is the spindle checkpoint. By contrast, progression through mitosis during the cell cycles of early embryos is largely checkpoint independent.7,8,9,10 Here, we first show that CDC-20 phosphoregulation controls mitotic duration in the C. elegans embryo and defines a checkpoint-independent temporal mitotic optimum for robust embryogenesis. CDC-20 phosphoregulation occurs at kinetochores and in the cytosol. At kinetochores, the flux of CDC-20 for local dephosphorylation requires an ABBA motif on BUB-1 that directly interfaces with the structured WD40 domain of CDC-20.6,11,12,13 We next show that a conserved "STP" motif in BUB-1 that docks the mitotic kinase PLK-114 is necessary for CDC-20 kinetochore recruitment and timely mitotic progression. The kinase activity of PLK-1 is required for CDC-20 to localize to kinetochores and phosphorylates the CDC-20-binding ABBA motif of BUB-1 to promote BUB-1-CDC-20 interaction and mitotic progression. Thus, the BUB-1-bound pool of PLK-1 ensures timely mitosis during embryonic cell cycles by promoting CDC-20 recruitment to the vicinity of kinetochore-localized phosphatase activity.
Collapse
Affiliation(s)
- Jack Houston
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Midori Ohta
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - J Sebastián Gómez-Cavazos
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Amar Deep
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kevin D Corbett
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karen Oegema
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Pablo Lara-Gonzalez
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Taekyung Kim
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; Department of Biology Education, Pusan National University, Busan 46241, Republic of Korea.
| | - Arshad Desai
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
48
|
Taylor SJP, Bel Borja L, Soubigou F, Houston J, Cheerambathur DK, Pelisch F. BUB-1 and CENP-C recruit PLK-1 to control chromosome alignment and segregation during meiosis I in C. elegans oocytes. eLife 2023; 12:e84057. [PMID: 37067150 PMCID: PMC10156168 DOI: 10.7554/elife.84057] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 04/14/2023] [Indexed: 04/18/2023] Open
Abstract
Phosphorylation is a key post-translational modification that is utilised in many biological processes for the rapid and reversible regulation of protein localisation and activity. Polo-like kinase 1 (PLK-1) is essential for both mitotic and meiotic cell divisions, with key functions being conserved in eukaryotes. The roles and regulation of PLK-1 during mitosis have been well characterised. However, the discrete roles and regulation of PLK-1 during meiosis have remained obscure. Here, we used Caenorhabditis elegans oocytes to show that PLK-1 plays distinct roles in meiotic spindle assembly and/or stability, chromosome alignment and segregation, and polar body extrusion during meiosis I. Furthermore, by a combination of live imaging and biochemical analysis we identified the chromosomal recruitment mechanisms of PLK-1 during C. elegans oocyte meiosis. The spindle assembly checkpoint kinase BUB-1 directly recruits PLK-1 to the kinetochore and midbivalent while the chromosome arm population of PLK-1 depends on a direct interaction with the centromeric-associated protein CENP-CHCP-4. We found that perturbing both BUB-1 and CENP-CHCP-4 recruitment of PLK-1 leads to severe meiotic defects, resulting in highly aneuploid oocytes. Overall, our results shed light on the roles played by PLK-1 during oocyte meiosis and provide a mechanistic understanding of PLK-1 targeting to meiotic chromosomes.
Collapse
Affiliation(s)
- Samuel JP Taylor
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Laura Bel Borja
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Flavie Soubigou
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Jack Houston
- Ludwig Institute for Cancer Research, San Diego BranchLa JollaUnited States
| | - Dhanya K Cheerambathur
- Wellcome Centre for Cell Biology & Institute of Cell Biology, School of Biological Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Federico Pelisch
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| |
Collapse
|
49
|
Dudka D, Akins RB, Lampson MA. FREEDA: an automated computational pipeline guides experimental testing of protein innovation by detecting positive selection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530329. [PMID: 36909479 PMCID: PMC10002610 DOI: 10.1101/2023.02.27.530329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Cell biologists typically focus on conserved regions of a protein, overlooking innovations that can shape its function over evolutionary time. Computational analyses can reveal potential innovations by detecting statistical signatures of positive selection that leads to rapid accumulation of beneficial mutations. However, these approaches are not easily accessible to non-specialists, limiting their use in cell biology. Here, we present an automated computational pipeline FREEDA (Finder of Rapidly Evolving Exons in De novo Assemblies) that provides a simple graphical user interface requiring only a gene name, integrates widely used molecular evolution tools to detect positive selection, and maps results onto protein structures predicted by AlphaFold. Applying FREEDA to >100 mouse centromere proteins, we find evidence of positive selection in intrinsically disordered regions of ancient domains, suggesting innovation of essential functions. As a proof-of-principle experiment, we show innovation in centromere binding of CENP-O. Overall, we provide an accessible computational tool to guide cell biology research and apply it to experimentally demonstrate functional innovation.
Collapse
|
50
|
Al-Rawi A, Kaye E, Korolchuk S, Endicott JA, Ly T. Cyclin A and Cks1 promote kinase consensus switching to non-proline-directed CDK1 phosphorylation. Cell Rep 2023; 42:112139. [PMID: 36840943 DOI: 10.1016/j.celrep.2023.112139] [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: 06/06/2022] [Revised: 11/17/2022] [Accepted: 02/02/2023] [Indexed: 02/26/2023] Open
Abstract
Ordered protein phosphorylation by CDKs is a key mechanism for regulating the cell cycle. How temporal order is enforced in mammalian cells remains unclear. Using a fixed cell kinase assay and phosphoproteomics, we show how CDK1 activity and non-catalytic CDK1 subunits contribute to the choice of substrate and site of phosphorylation. Increases in CDK1 activity alter substrate choice, with intermediate- and low-sensitivity CDK1 substrates enriched in DNA replication and mitotic functions, respectively. This activity dependence is shared between Cyclin A- and Cyclin B-CDK1. Cks1 has a proteome-wide role as an enhancer of multisite CDK1 phosphorylation. Contrary to the model of CDK1 as an exclusively proline-directed kinase, we show that Cyclin A and Cks1 enhance non-proline-directed phosphorylation, preferably on sites with a +3 lysine residue. Indeed, 70% of cell-cycle-regulated phosphorylations, where the kinase carrying out this modification has not been identified, are non-proline-directed CDK1 sites.
Collapse
Affiliation(s)
- Aymen Al-Rawi
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Edward Kaye
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | | | - Jane A Endicott
- Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Tony Ly
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK.
| |
Collapse
|