1
|
Tang W, Li J, Zhou Y, Li J, Ma Z, Li X, Wang H, Xiong M, Chen X, Li X, Chen W, Ma H, Ye X. Palmatine attenuates MYH9 mediated nuclear localization of AURKA to induce G2/M phase arrest in colorectal cancer cells. Int Immunopharmacol 2024; 143:113615. [PMID: 39536490 DOI: 10.1016/j.intimp.2024.113615] [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: 08/27/2024] [Revised: 10/26/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The mitotic kinase Aurora kinase A (AURKA), which plays a crucial role in cell cycle progression, represents a promising target for the treatment of colorectal cancer (CRC). Here, we found that AURKA is a target of a CRC suppressor, the Palmatine (PAL). However, the underlying mechanism remains elusive. This work aims to investigate the underlying mechanism how PAL suppresses CRC through AURKA. It was confirmed that AURKA played an important role in the development of CRC tumors through an Azoxymethane/Dextran sulfate sodium salt induced mice model and tissue microarrays of CRC-patients. Overexpression of AURKA was able to partially reverse the inhibitory effect of PAL on CRC cells, showing that PAL significantly inhibited the malignant phenotype and induced the G2/M phase arrest of CRC cells by down-regulating AURKA. Functional studies indicated that PAL attenuated the stability of AURKA protein and reduced its nuclear level, resulting in reduction of key proteins in the G2/M phase. Importantly, Co-IP and WB experiments suggested that Myosin heavy chain 9 (MYH9) interacted with AURKA and had an impact on its nuclear localization. PAL can decrease nuclear AURKA by reducing the interaction of AURKA and MYH9. Taken together, our study revealed that MYH9 as an auxiliary protein for the nuclear localization of AURKA and elucidated the mechanism that PAL reduced nuclear AURKA through inhibiting the interaction of AURKA and MYH9 to induce G2/M phase arrest in CRC cells. Therefore, this study may provide a theoretical basis of PAL for the treatment of CRC.
Collapse
Affiliation(s)
- Wanyu Tang
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Jingwei Li
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Yuan Zhou
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Juan Li
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Xiaoduo Li
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Hongmei Wang
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Mengyuan Xiong
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Xiantao Chen
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Xuegang Li
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China.
| | - Hang Ma
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
2
|
Deshpande RP, Wu K, Wu SY, Tyagi A, Smith EC, Hunting J, Ruiz J, Li W, Watabe K. Tumor-intrinsic CDC42BPB confers resistance to anti-PD-1 immune checkpoint blockade in breast cancer. Mol Ther 2024; 32:3669-3682. [PMID: 39086134 PMCID: PMC11489557 DOI: 10.1016/j.ymthe.2024.07.021] [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: 01/24/2024] [Revised: 06/04/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024] Open
Abstract
Immune checkpoint blockade has been used to treat breast cancer, but the clinical responses remain relatively poor. We have used the CRISPR-Cas9 kinome knockout library consisting of 763 kinase genes to identify tumor-intrinsic kinases conferring resistance to anti-PD-1 immune checkpoint blockade. We have identified the CDC42BPB kinase as a potential target to overcome the resistance to anti-PD-1 immune checkpoint blockade immunotherapy. We found that CDC42BPB is highly expressed in breast cancer patients who are non-responsive to immunotherapy. Furthermore, a small-molecule pharmacological inhibitor, BDP5290, which targets CDC42BPB, synergized with anti-PD-1 and enhanced tumor cell killing by promoting T cell proliferation in both in vitro and in vivo assays. Moreover, anti-PD-1-resistant breast cancer cells showed higher expression of CDC42BPB, and its inhibition rendered the resistant cells more susceptible to T cell killing in the presence of anti-PD-1. We also found that CDC42BPB phosphorylated AURKA, which in turn upregulated PD-L1 through cMYC. Our results have revealed a robust link between tumor-intrinsic kinase and immunotherapy resistance and have provided a rationale for a unique combination therapy of CDC42BPB inhibition and anti-PD-1 immunotherapy for breast cancer.
Collapse
Affiliation(s)
- Ravindra Pramod Deshpande
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Kerui Wu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Abhishek Tyagi
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Eleanor C Smith
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - John Hunting
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Jimmy Ruiz
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Wencheng Li
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
3
|
Li Y, Wu M, Liu Y, Sun L, Mu P, Ma B, Xie J. Haspin mediates H3.3S31 phosphorylation downstream of Aurora B in mouse embryonic stem cells. Protein Sci 2024; 33:e5126. [PMID: 39073155 PMCID: PMC11284449 DOI: 10.1002/pro.5126] [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: 01/09/2024] [Revised: 07/09/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024]
Abstract
Histone phosphorylation is instrumental in regulating diverse cellular processes across eukaryotes. Unraveling the kinases that target specific histone sites is key to deciphering the underlying mechanisms. Among the various sites on histone tails that can undergo phosphorylation, the kinase responsible for H3.3S31 phosphorylation remained elusive. Since both H3.3S31ph and H3T3ph occur specifically during mitosis, and Haspin is the known kinase for H3T3 phosphorylation, we investigated its potential role in H3.3S31 phosphorylation. We employed CRISPR/Cas9, RNA interference, and specific small molecule inhibitors to eliminate Haspin function in various cell types. Our data consistently revealed a link between Haspin and H3.3S31ph. Furthermore, in vitro kinase assays provided evidence supporting Haspin's contribution to H3.3S31ph. Loss- and gain-of-function experiments targeting Haspin and Aurora B further suggested a hierarchical relationship. Haspin acts as a downstream kinase of Aurora B, specifically orchestrating H3.3S31 phosphorylation in mESCs. This study unveils a novel role for Haspin as a kinase in regulating H3.3S31 phosphorylation during mitosis. This discovery holds promise for expanding our understanding of the functional significance of Haspin and H3.3S31ph in mammals.
Collapse
Affiliation(s)
- Yuanyuan Li
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- The Center for Reproductive Medicine, Shanghai East HospitalTongji UniversityShanghaiChina
| | - Meixian Wu
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yang Liu
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Lihua Sun
- The Center for Reproductive Medicine, Shanghai East HospitalTongji UniversityShanghaiChina
| | - Peiqiang Mu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life SciencesSouth China Agricultural UniversityGuangzhouChina
| | - Binbin Ma
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Present address:
Department of BiologyThe Johns Hopkins UniversityBaltimoreMarylandUSA
| | - Jing Xie
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- The Center for Reproductive Medicine, Shanghai East HospitalTongji UniversityShanghaiChina
| |
Collapse
|
4
|
Shawky MM, Abdallah M, Khalifa H, Aboushady Y, Abadi AH, Engel M, Abdel-Halim M. Synthesis and evaluation of novel N1-acylated 5-(4-pyridinyl)indazole derivatives as potent and selective haspin inhibitors. Bioorg Chem 2024; 145:107235. [PMID: 38447464 DOI: 10.1016/j.bioorg.2024.107235] [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: 01/10/2024] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
Protein kinase dysregulation was strongly linked to cancer pathogenesis. Moreover, histone alterations were found to be among the most important post-translational modifications that could contribute to cancer growth and development. In this context, haspin, an atypical serine/threonine kinase, phosphorylates histone H3 at threonine-3 and is notably overexpressed in various common cancer types. Herein, we report novel 5-(4-pyridinyl)indazole derivatives as potent and selective haspin inhibitors. Amide coupling at N1 of the indazole ring with m-hydroxyphenyl acetic acid yielded compound 21 with an IC50 value of 78 nM against haspin. This compound showed a meaningful selectivity over 15 of the most common off-targets, including Clk 1-3 and Dyrk1A, 1B, and 2. The most potent haspin inhibitors 5 and 21 effectively inhibited the growth of the NCI-60 cancer cell lines, further emphasizing the success of our scaffold as a new selective lead for the development of anti-cancer therapeutic agents.
Collapse
Affiliation(s)
- Mona M Shawky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Mennatallah Abdallah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Hend Khalifa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Youssef Aboushady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123 Saarbrücken, Germany.
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt.
| |
Collapse
|
5
|
Chu S, Xie X, Payan C, Stochaj U. Valosin containing protein (VCP): initiator, modifier, and potential drug target for neurodegenerative diseases. Mol Neurodegener 2023; 18:52. [PMID: 37545006 PMCID: PMC10405438 DOI: 10.1186/s13024-023-00639-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/27/2023] [Indexed: 08/08/2023] Open
Abstract
The AAA+ ATPase valosin containing protein (VCP) is essential for cell and organ homeostasis, especially in cells of the nervous system. As part of a large network, VCP collaborates with many cofactors to ensure proteostasis under normal, stress, and disease conditions. A large number of mutations have revealed the importance of VCP for human health. In particular, VCP facilitates the dismantling of protein aggregates and the removal of dysfunctional organelles. These are critical events to prevent malfunction of the brain and other parts of the nervous system. In line with this idea, VCP mutants are linked to the onset and progression of neurodegeneration and other diseases. The intricate molecular mechanisms that connect VCP mutations to distinct brain pathologies continue to be uncovered. Emerging evidence supports the model that VCP controls cellular functions on multiple levels and in a cell type specific fashion. Accordingly, VCP mutants derail cellular homeostasis through several mechanisms that can instigate disease. Our review focuses on the association between VCP malfunction and neurodegeneration. We discuss the latest insights in the field, emphasize open questions, and speculate on the potential of VCP as a drug target for some of the most devastating forms of neurodegeneration.
Collapse
Affiliation(s)
- Siwei Chu
- Department of Physiology, McGill University, Montreal, HG3 1Y6, Canada
| | - Xinyi Xie
- Department of Physiology, McGill University, Montreal, HG3 1Y6, Canada
| | - Carla Payan
- Department of Physiology, McGill University, Montreal, HG3 1Y6, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, HG3 1Y6, Canada.
- Quantitative Life Sciences Program, McGill University, Montreal, Canada.
| |
Collapse
|
6
|
Cheng A, Xu T, You W, Wang T, Zhang D, Guo H, Zhang H, Pan X, Wang Y, Liu L, Zhang K, Shi J, Yao X, Guo J, Yang Z. A mitotic NADPH upsurge promotes chromosome segregation and tumour progression in aneuploid cancer cells. Nat Metab 2023; 5:1141-1158. [PMID: 37349486 DOI: 10.1038/s42255-023-00832-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 05/26/2023] [Indexed: 06/24/2023]
Abstract
Redox metabolites have been observed to fluctuate through the cell cycle in cancer cells, but the functional impacts of such metabolic oscillations remain unknown. Here, we uncover a mitosis-specific nicotinamide adenine dinucleotide phosphate (NADPH) upsurge that is essential for tumour progression. Specifically, NADPH is produced by glucose 6-phosphate dehydrogenase (G6PD) upon mitotic entry, which neutralizes elevated reactive oxygen species (ROS) and prevents ROS-mediated inactivation of mitotic kinases and chromosome missegregation. Mitotic activation of G6PD depends on the phosphorylation of its co-chaperone protein BAG3 at threonine 285, which results in dissociation of inhibitory BAG3. Blocking BAG3T285 phosphorylation induces tumour suppression. A mitotic NADPH upsurge is present in aneuploid cancer cells with high levels of ROS, while nearly unobservable in near-diploid cancer cells. High BAG3T285 phosphorylation is associated with worse prognosis in a cohort of patients with microsatellite-stable colorectal cancer. Our study reveals that aneuploid cancer cells with high levels of ROS depend on a G6PD-mediated NADPH upsurge in mitosis to protect them from ROS-induced chromosome missegregation.
Collapse
Affiliation(s)
- Aoxing Cheng
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tian Xu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weiyi You
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ting Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dongming Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huimin Guo
- Center for Biological Technology, Anhui Agricultural University, Hefei, China
| | - Haiyan Zhang
- Core Facility Centre for Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin Pan
- National Center of Biomedical Analysis of China, Beijing, China
| | - Yucai Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Liu Liu
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jue Shi
- Center for Quantitative Systems Biology, Department of Physics and Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Guo
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Zhenye Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
7
|
Lin CI, Chen ZC, Chen CH, Chang YH, Lee TC, Tang TT, Yu TW, Yang CM, Tsai MC, Huang CC, Yang TW, Lin CC, Wang RH, Chiou GY, Jong YJ, Chao JI. Co-inhibition of Aurora A and Haspin kinases enhances survivin blockage and p53 induction for mitotic catastrophe and apoptosis in human colorectal cancer. Biochem Pharmacol 2022; 206:115289. [PMID: 36241092 DOI: 10.1016/j.bcp.2022.115289] [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: 07/07/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is a leading cause and mortality worldwide. Aurora A and haspin kinases act pivotal roles in mitotic progression. However, the blockage of Aurora A and Haspin for CRC therapy is still unclear. Here we show that the Haspin and p-H3T3 protein levels were highly expressed in CRC tumor tissues of clinical patients. Overexpression of Haspin increased the protein levels of p-H3T3 and survivin in human CRC cells; conversely, the protein levels of p-H3T3 and survivin were decreased by the Haspin gene knockdown. Moreover, the gene knockdown of Aurora A induced abnormal chromosome segregation, mitotic catastrophe, and cell growth inhibition. Combined targeted by co-treatment of CHR6494, a Haspin inhibitor, and MLN8237, an Aurora A inhibitor, enhanced apoptosis and CRC tumor inhibition. MLN8237 and CHR6494 induced abnormal chromosome segregation and mitotic catastrophe. Meanwhile, MLN8237 and CHR6494 inhibited survivin protein levels but conversely induced p53 protein expression. Ectopic survivin expression by transfection with a survivin-expressed vector resisted the cell death in the MLN8237- and CHR6494-treated cells. In contrast, the existence of functional p53 increased the apoptotic levels by treatment with MLN8237 and CHR6494. Co-treatment of CHR6494 and MLN8237 enhanced the blockage of human CRC xenograft tumors in nude mice. Taken together, co-inhibition of Aurora A and Haspin enhances survivin inhibition, p53 pathway induction, mitotic catastrophe, apoptosis and tumor inhibition that may provide a potential strategy for CRC therapy.
Collapse
Affiliation(s)
- Chien-I Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zan-Chu Chen
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Chien-Hung Chen
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yun-Hsuan Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tsai-Chia Lee
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tsai-Tai Tang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tzu-Wei Yu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Chih-Man Yang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Ming-Chang Tsai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chi-Chou Huang
- Division of Colon and Rectum, Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tzu-Wei Yang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Che Lin
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Rou-Hsin Wang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Guang-Yuh Chiou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yuh-Jyh Jong
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Departments of Pediatrics and Laboratory Medicine, and Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Jui-I Chao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan; Center For Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| |
Collapse
|
8
|
Liu Y, Yang H, Fang Y, Xing Y, Pang X, Li Y, Zhang Y, Liu Y. Function and inhibition of Haspin kinase: targeting multiple cancer therapies by antimitosis. J Pharm Pharmacol 2022; 75:445-465. [PMID: 36334086 DOI: 10.1093/jpp/rgac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
Abstract
Objectives
Haploid germ cell-specific nuclear protein kinase (Haspin) is a serine/threonine kinase as an atypical kinase, which is structurally distinct from conventional protein kinases.
Key findings
Functionally, Haspin is involved in important cell cycle progression, particularly in critical mitosis regulating centromeric sister chromatid cohesion during prophase and prometaphase, and subsequently ensuring proper chromosome alignment during metaphase and the normal chromosome segregation during anaphase. However, increasing evidence has demonstrated that Haspin is significantly upregulated in a variety of cancer cells in addition to normal proliferating somatic cells. Its knockdown or small molecule inhibition could prevent cancer cell growth and induce apoptosis by disrupting the regular mitotic progression. Given the specificity of its expressed tissues or cells and the uniqueness of its current known substrate, Haspin can be a promising target against cancer. Consequently, selective synthetic and natural inhibitors of Haspin have been widely developed to determine their inhibitory power for various cancer cells in vivo and in vitro.
Summary
Here our perspective includes a comprehensive review of the roles and structure of Haspin, its relatively potent and selective inhibitors and Haspin’s preliminary studies in a variety of cancers.
Collapse
Affiliation(s)
- Yongjian Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Hongliu Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yongsheng Fang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yantao Xing
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Xinxin Pang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yuanyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yonggang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| |
Collapse
|
9
|
Jiang Y, Wang T, Sheng D, Han C, Xu T, Zhang P, You W, Fan W, Zhang Z, Jin T, Duan X, Yuan X, Liu X, Zhang K, Ruan K, Shi J, Guo J, Cheng A, Yang Z. Aurora A-mediated pyruvate kinase M2 phosphorylation promotes biosynthesis with glycolytic metabolites and tumor cell cycle progression. J Biol Chem 2022; 298:102561. [PMID: 36198360 PMCID: PMC9637814 DOI: 10.1016/j.jbc.2022.102561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer cells have distinctive demands for intermediates from glucose metabolism for biosynthesis and energy in different cell cycle phases. However, how cell cycle regulators and glycolytic enzymes coordinate to orchestrate the essential metabolic processes are still poorly characterized. Here, we report a novel interaction between the mitotic kinase, Aurora A, and the glycolytic enzyme, pyruvate kinase M2 (PKM2), in the interphase of the cell cycle. We found Aurora A–mediated phosphorylation of PKM2 at threonine 45. This phosphorylation significantly attenuated PKM2 enzymatic activity by reducing its tetramerization and also promoted glycolytic flux and the branching anabolic pathways. Replacing the endogenous PKM2 with a nonphosphorylated PKM2 T45A mutant inhibited glycolysis, glycolytic branching pathways, and tumor growth in both in vitro and in vivo models. Together, our study revealed a new protumor function of Aurora A through modulating a rate-limiting glycolytic enzyme, PKM2, mainly during the S phase of the cell cycle. Our findings also showed that although both Aurora A and Aurora B kinase phosphorylate PKM2 at the same residue, the spatial and temporal regulations of the specific kinase and PKM2 interaction are context dependent, indicating intricate interconnectivity between cell cycle and glycolytic regulators.
Collapse
Affiliation(s)
- Ya Jiang
- Department of Digestive Disease, the First affiliated hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China; MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Ting Wang
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Dandan Sheng
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Chaoqiang Han
- Department of Digestive Disease, the First affiliated hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China
| | - Tian Xu
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Peng Zhang
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Weiyi You
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Weiwei Fan
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Zhiyong Zhang
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Digestive Disease, the First affiliated hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China
| | - Xiaotao Duan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiao Yuan
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology &CAS Center of Excellence in Molecular Cell Sciences, Hefei, Anhui 230026, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology &CAS Center of Excellence in Molecular Cell Sciences, Hefei, Anhui 230026, China
| | - Kaiguang Zhang
- Department of Digestive Disease, the First affiliated hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China
| | - Ke Ruan
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China
| | - Jue Shi
- Center for Quantitative Systems Biology, Department of Physics and Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Jing Guo
- Department of Digestive Disease, the First affiliated hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China
| | - Aoxing Cheng
- Department of Digestive Disease, the First affiliated hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China.
| | - Zhenye Yang
- Department of Digestive Disease, the First affiliated hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China; MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei 230027, China.
| |
Collapse
|
10
|
Dissecting the roles of Haspin and VRK1 in histone H3 phosphorylation during mitosis. Sci Rep 2022; 12:11210. [PMID: 35778595 PMCID: PMC9249732 DOI: 10.1038/s41598-022-15339-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/22/2022] [Indexed: 12/12/2022] Open
Abstract
Protein kinases that phosphorylate histones are ideally-placed to influence the behavior of chromosomes during cell division. Indeed, a number of conserved histone phosphorylation events occur prominently during mitosis and meiosis in most eukaryotes, including on histone H3 at threonine-3 (H3T3ph). At least two kinases, Haspin and VRK1 (NHK-1/ballchen in Drosophila), have been proposed to carry out this modification. Phosphorylation of H3 by Haspin has defined roles in mitosis, but the significance of VRK1 activity towards histones in dividing cells has been unclear. Here, using in vitro kinase assays, KiPIK screening, RNA interference, and CRISPR/Cas9 approaches, we were unable to substantiate a direct role for VRK1, or its paralogue VRK2, in the phosphorylation of threonine-3 or serine-10 of Histone H3 in mitosis, although loss of VRK1 did slow cell proliferation. We conclude that the role of VRKs, and their more recently identified association with neuromuscular disease and importance in cancers of the nervous system, are unlikely to involve mitotic histone kinase activity. In contrast, Haspin is required to generate H3T3ph during mitosis.
Collapse
|
11
|
HASPIN kinase inhibitor CHR-6494 suppresses intestinal polyp development, cachexia, and hypogonadism in Apcmin/+ mice. Eur J Cancer Prev 2021; 29:481-485. [PMID: 31833958 PMCID: PMC7531494 DOI: 10.1097/cej.0000000000000562] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
HASPIN has been identified as a nuclear Ser/Thr kinase specifically expressed in haploid germ cells. HASPIN kinase inhibitors were recently isolated, and their antitumor activity reported. Colorectal cancer occurs with high incidence worldwide. In this study, we examined whether HASPIN inhibitor CHR-6494 suppresses cancer progression in ApcMin/+ mice, a familial colon tumor disease model. Mice were treated by intraperitoneal injection of CHR-6494 for 50 days. Following the treatment period, intestinal polyps were counted and testosterone and spermatogenesis levels were observed. Intraperitoneal administration of CHR-6494 significantly inhibited intestinal polyp development and recovered body weight in ApcMin/+ mice. Although spermatogenesis was inhibited with increasing age in ApcMin/+ mice, CHR-6494 significantly improved blood testosterone levels and spermatogenesis. Our results suggest that HASPIN inhibitors may be useful as anti-cancer agents and for the treatment of hypogonadism in colorectal cancer patients.
Collapse
|
12
|
Vichas A, Riley AK, Nkinsi NT, Kamlapurkar S, Parrish PCR, Lo A, Duke F, Chen J, Fung I, Watson J, Rees M, Gabel AM, Thomas JD, Bradley RK, Lee JK, Hatch EM, Baine MK, Rekhtman N, Ladanyi M, Piccioni F, Berger AH. Integrative oncogene-dependency mapping identifies RIT1 vulnerabilities and synergies in lung cancer. Nat Commun 2021; 12:4789. [PMID: 34373451 PMCID: PMC8352964 DOI: 10.1038/s41467-021-24841-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
CRISPR-based cancer dependency maps are accelerating advances in cancer precision medicine, but adequate functional maps are limited to the most common oncogenes. To identify opportunities for therapeutic intervention in other rarer subsets of cancer, we investigate the oncogene-specific dependencies conferred by the lung cancer oncogene, RIT1. Here, genome-wide CRISPR screening in KRAS, EGFR, and RIT1-mutant isogenic lung cancer cells identifies shared and unique vulnerabilities of each oncogene. Combining this genetic data with small-molecule sensitivity profiling, we identify a unique vulnerability of RIT1-mutant cells to loss of spindle assembly checkpoint regulators. Oncogenic RIT1M90I weakens the spindle assembly checkpoint and perturbs mitotic timing, resulting in sensitivity to Aurora A inhibition. In addition, we observe synergy between mutant RIT1 and activation of YAP1 in multiple models and frequent nuclear overexpression of YAP1 in human primary RIT1-mutant lung tumors. These results provide a genome-wide atlas of oncogenic RIT1 functional interactions and identify components of the RAS pathway, spindle assembly checkpoint, and Hippo/YAP1 network as candidate therapeutic targets in RIT1-mutant lung cancer.
Collapse
Affiliation(s)
- Athea Vichas
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Amanda K Riley
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Naomi T Nkinsi
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shriya Kamlapurkar
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Phoebe C R Parrish
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - April Lo
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Fujiko Duke
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Jennifer Chen
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Iris Fung
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | | | - Matthew Rees
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Austin M Gabel
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - James D Thomas
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Robert K Bradley
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - John K Lee
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Emily M Hatch
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marina K Baine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Federica Piccioni
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Merck Research Laboratories, Boston, MA, USA
| | - Alice H Berger
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
13
|
Wang R, Ascanelli C, Abdelbaki A, Fung A, Rasmusson T, Michaelides I, Roberts K, Lindon C. Selective targeting of non-centrosomal AURKA functions through use of a targeted protein degradation tool. Commun Biol 2021; 4:640. [PMID: 34050235 PMCID: PMC8163823 DOI: 10.1038/s42003-021-02158-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/29/2021] [Indexed: 12/22/2022] Open
Abstract
Targeted protein degradation tools are becoming a new therapeutic modality, allowing small molecule ligands to be reformulated as heterobifunctional molecules (PROteolysis Targeting Chimeras, PROTACs) that recruit ubiquitin ligases to targets of interest, leading to ubiquitination and destruction of the targets. Several PROTACs against targets of clinical interest have been described, but detailed descriptions of the cell biology modulated by PROTACs are missing from the literature. Here we describe the functional characterization of a PROTAC derived from AURKA inhibitor MLN8237 (alisertib). We demonstrate efficient and specific destruction of both endogenous and overexpressed AURKA by Cereblon-directed PROTACs. At the subcellular level, we find differential targeting of AURKA on the mitotic spindle compared to centrosomes. The phenotypic consequences of PROTAC treatment are therefore distinct from those mediated by alisertib, and in mitotic cells differentially regulate centrosome- and chromatin- based microtubule spindle assembly pathways. In interphase cells PROTAC-mediated clearance of non-centrosomal AURKA modulates the cytoplasmic role played by AURKA in mitochondrial dynamics, whilst the centrosomal pool is refractory to PROTAC-mediated clearance. Our results point to differential sensitivity of subcellular pools of substrate, governed by substrate conformation or localization-dependent accessibility to PROTAC action, a phenomenon not previously described for this new class of degrader compounds. Wang et al develop tools to target the mitotic regulator AURKA by synthesising PROTACs based on the inhibitor MLN8237. They find that the new PROTAC compound efficiently clears cytoplasmic and mitotic spindle-associated AURKA but does not eliminate AURKA activity from centrosomes, demonstrating the possibility of targeting subpopulations.
Collapse
Affiliation(s)
- Richard Wang
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Ahmed Abdelbaki
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Alex Fung
- Department of Pharmacology, University of Cambridge, Cambridge, UK.,University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Tim Rasmusson
- Discovery Sciences, R&D, AstraZeneca, Cambridge, UK.,Bristol Myers Squibb, Cambridge, MA, USA
| | | | | | - Catherine Lindon
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
14
|
Cheng A, Jiang Y, Wang T, Yu F, Ishrat I, Zhang D, Ji X, Chen M, Xiao W, Li Q, Zhang K, Niu G, Shi J, Pan Y, Yang Z, Guo J. Energy restriction causes metaphase delay and chromosome mis-segregation in cancer cells. Cell Cycle 2021; 20:1195-1208. [PMID: 34048314 DOI: 10.1080/15384101.2021.1930679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
ATP metabolism during mitosis needs to be coordinated with numerous energy-demanding activities, especially in cancer cells whose metabolic pathways are reprogramed to sustain rapid proliferation in a nutrient-deficient environment. Although strategies targeting the energy metabolic pathways have shown therapeutic efficacy in preclinical cancer models, how normal cells and cancer cells differentially respond to energy shortage is unclear. In this study, using time-lapse microscopy, we found that cancer cells displayed unique mitotic phenotypes in a dose-dependent manner upon decreasing ATP (i.e. energy) supply. When reduction in ATP concentration was moderate, chromosome movements in mitosis were barely affected, while the metaphase-anaphase transition was significantly prolonged due to reduced tension between the sister-kinetochores, which delayed the satisfaction of the spindle assembly checkpoint. Further reduction in ATP concentration led to a decreased level of Aurora-B at the centromere, resulting in increased chromosome mis-segregation after metaphase delay. In contrast to cancer cells, ATP restriction in non-transformed cells induced cell cycle arrest in interphase, rather than causing mitotic defects. In addition, data mining of cancer patient database showed a correlation between signatures of energy production and chromosomal instability possibly resulted from mitotic defects. Together, these results reveal that energy restriction induces differential responses in normal and cancer cells, with chromosome mis-segregation only observed in cancer cells. This points to targeting energy metabolism as a potentially cancer-selective therapeutic strategy.
Collapse
Affiliation(s)
- Aoxing Cheng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ya Jiang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ting Wang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fazhi Yu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Iqra Ishrat
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dongming Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaoyang Ji
- Joint Turing-Darwin Laboratory of Phil Rivers Technology Ltd. And Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.,Department of Computational Biology, Phil Rivers Technology Ltd, Beijing, China
| | - Minhua Chen
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weihua Xiao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qing Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Gang Niu
- Joint Turing-Darwin Laboratory of Phil Rivers Technology Ltd. And Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.,Department of Computational Biology, Phil Rivers Technology Ltd, Beijing, China.,West Institute of Computing Technology, Chinese Academy of Sciences, Chongqing, China
| | - Jue Shi
- Center for Quantitative Systems Biology, Department of Physics and Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Yueyin Pan
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhenye Yang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,University of Science and Technology of China, Hefei, China
| | - Jing Guo
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
15
|
Nadarajan S, Altendorfer E, Saito TT, Martinez-Garcia M, Colaiácovo MP. HIM-17 regulates the position of recombination events and GSP-1/2 localization to establish short arm identity on bivalents in meiosis. Proc Natl Acad Sci U S A 2021; 118:e2016363118. [PMID: 33883277 PMCID: PMC8092412 DOI: 10.1073/pnas.2016363118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The position of recombination events established along chromosomes in early prophase I and the chromosome remodeling that takes place in late prophase I are intrinsically linked steps of meiosis that need to be tightly regulated to ensure accurate chromosome segregation and haploid gamete formation. Here, we show that RAD-51 foci, which form at the sites of programmed meiotic DNA double-strand breaks (DSBs), exhibit a biased distribution toward off-centered positions along the chromosomes in wild-type Caenorhabditis elegans, and we identify two meiotic roles for chromatin-associated protein HIM-17 that ensure normal chromosome remodeling in late prophase I. During early prophase I, HIM-17 regulates the distribution of DSB-dependent RAD-51 foci and crossovers on chromosomes, which is critical for the formation of distinct chromosome subdomains (short and long arms of the bivalents) later during chromosome remodeling. During late prophase I, HIM-17 promotes the normal expression and localization of protein phosphatases GSP-1/2 to the surface of the bivalent chromosomes and may promote GSP-1 phosphorylation, thereby antagonizing Aurora B kinase AIR-2 loading on the long arms and preventing premature loss of sister chromatid cohesion. We propose that HIM-17 plays distinct roles at different stages during meiotic progression that converge to promote normal chromosome remodeling and accurate chromosome segregation.
Collapse
Affiliation(s)
| | - Elisabeth Altendorfer
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Takamune T Saito
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | | | - Monica P Colaiácovo
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
16
|
Chen A, Wen S, Liu F, Zhang Z, Liu M, Wu Y, He B, Yan M, Kang T, Lam EWF, Wang Z, Liu Q. CRISPR/Cas9 screening identifies a kinetochore-microtubule dependent mechanism for Aurora-A inhibitor resistance in breast cancer. Cancer Commun (Lond) 2021; 41:121-139. [PMID: 33471959 PMCID: PMC7896750 DOI: 10.1002/cac2.12125] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 12/31/2022] Open
Abstract
Background Overexpression of Aurora‐A (AURKA) is a feature of breast cancer and associates with adverse prognosis. The selective Aurora‐A inhibitor alisertib (MLN8237) has recently demonstrated promising antitumor responses as a single agent in various cancer types but its phase III clinical trial was reported as a failure since MLN8237 did not show an apparent effect in prolonging the survival of patients. Thus, identification of potential targets that could enhance the activity of MLN8237 would provide a rationale for drug combination to achieve better therapeutic outcome. Methods Here, we conducted a systematic synthetic lethality CRISPR/Cas9 screening of 507 kinases using MLN8237 in breast cancer cells and identified a number of targetable kinases that displayed synthetic lethality interactions with MLN8237. Then, we performed competitive growth assays, colony formation assays, cell viability assays, apoptosis assays, and xenograft murine model to evaluate the synergistic therapeutic effects of Haspin (GSG2) depletion or inhibition with MLN8237. For mechanistic studies, immunofluorescence was used to detect the state of microtubules and the localization of Aurora‐B and mitotic centromere‐associated kinesin (MCAK). Results Among the hits, we observed that Haspin depletion or inhibition marginally inhibited breast cancer cell growth but could substantially enhance the killing effects of MLN8237. Mechanistic studies showed that co‐treatment with Aurora‐A and Haspin inhibitors abolished the recruitment of Aurora‐B and mitotic centromere‐associated kinesin (MCAK) to centromeres which were associated with excessive microtubule depolymerization, kinetochore‐microtubule (KT‐MT) attachment failure, and severe mitotic catastrophe. We further showed that the combination of MLN8237 and the Haspin inhibitor CHR‐6494 synergistically reduced breast cancer cell viability and significantly inhibited both in vitro and in vivo tumor growth. Conclusions These findings establish Haspin as a synthetic lethal target and demonstrate CHR‐6494 as a potential combinational drug for promoting the therapeutic effects of MLN8237 on breast cancer.
Collapse
Affiliation(s)
- Ailin Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Shijun Wen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Fang Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Zijian Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Meiling Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Yuanzhong Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Bin He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Min Yan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Tiebang Kang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Eric W-F Lam
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China.,Department of Surgery and Cancer, Imperial College London, W12 0NN, London, UK
| | - Zifeng Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Quentin Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China.,Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, 116044, P. R. China
| |
Collapse
|
17
|
Huang Y, Liu Y, Zhu K, Ma X, Lu R, Zhang M. GSG2 Promotes Development and Predicts Poor Prognosis of Ovarian Cancer. Cancer Manag Res 2021; 13:499-508. [PMID: 33500663 PMCID: PMC7826093 DOI: 10.2147/cmar.s274807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/17/2020] [Indexed: 11/30/2022] Open
Abstract
Purpose Ovarian cancer is one of the most common malignant tumors in gynecology, whose treatment was seriously limited by the unclear understanding of molecular mechanism in disease development. GSG2, also known as Haspin, is a novel molecule found to be involved in human cancers. Materials and Methods In this study, immunohistochemical analysis was used to detect GSG2 expression in ovarian cancer tissues and corresponding normal tissues. Statistical analysis was performed to construct relationship between GSG2 and tumor characteristics as well as prognosis. Ovarian cell model with GSG2 knockdown was constructed through lentivirus-mediated transfection of shRNA, which was used in MTT assay, colony formation assay and flow cytometry for investigating the role of GSG2 in ovarian cancer. A human apoptosis antibody array was used to identify potential downstream apoptosis-related proteins of GSG2. Results The results demonstrated the upregulation of GSG2 in ovarian cancer, whose expression was positively related to tumor grade and AJCC stage, and negatively correlated with patients’ prognosis. Moreover, knockdown of GSG2 inhibited ovarian cancer development through suppressing cell growth and inducing cell apoptosis. Further exploration revealed that a variety of apoptosis-related and PI3K signaling pathway-related proteins may be implicated in the GSG2 induced regulation of ovarian cancer. Conclusion In summary, it was illustrated that GSG2 was involved in the development of ovarian cancer, which has the potential to become therapeutic target and prognostic indicator in ovarian cancer treatment.
Collapse
Affiliation(s)
- Yan Huang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yixuan Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Keyu Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Xiaolu Ma
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Renquan Lu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Meiqin Zhang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Huang M, Feng X, Su D, Wang G, Wang C, Tang M, Paulucci-Holthauzen A, Hart T, Chen J. Genome-wide CRISPR screen uncovers a synergistic effect of combining Haspin and Aurora kinase B inhibition. Oncogene 2020; 39:4312-4322. [PMID: 32300176 PMCID: PMC7291820 DOI: 10.1038/s41388-020-1296-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022]
Abstract
Aurora kinases are a family of serine/threonine kinases vital for cell division. Because of the overexpression of Aurora kinases in a broad range of cancers and their important roles in mitosis, inhibitors targeting Aurora kinases have attracted attention in cancer therapy. VX-680 is an effective pan-Aurora kinase inhibitor; however, its clinical efficacy was not satisfying. In this study, we performed CRISPR/Cas9 screens to identify genes whose depletion shows synthetic lethality with VX-680. The top hit from these screens was GSG2 (also known as Haspin), a serine/threonine kinase that phosphorylates histone H3 at Thr-3 during mitosis. Moreover, both Haspin knockout and Haspin inhibitor-treated HCT116 cells were hypersensitive to VX-680. Furthermore, we showed that the synthetic lethal interaction between Haspin depletion and VX-680 was mediated by the inhibition of Haspin with Aurora kinase B (AURKB), but not with Aurora kinase A (AURKA). Strikingly, combined inhibition of Haspin and AURKB had a better efficacy than single-agent treatment in both head and neck squamous cell carcinoma and non-small cell lung cancer. Taken together, our findings have uncovered a synthetic lethal interaction between AURKB and Haspin, which provides a strong rationale for this combination therapy for cancer patients.
Collapse
Affiliation(s)
- Min Huang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xu Feng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dan Su
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gang Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chao Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mengfan Tang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Traver Hart
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Cheng A, Zhang P, Wang B, Yang D, Duan X, Jiang Y, Xu T, Jiang Y, Shi J, Ding C, Wu G, Sang Z, Wu Q, Wang H, Wu M, Zhang Z, Pan X, Pan YY, Gao P, Zhang H, Zhou CZ, Guo J, Yang Z. Aurora-A mediated phosphorylation of LDHB promotes glycolysis and tumor progression by relieving the substrate-inhibition effect. Nat Commun 2019; 10:5566. [PMID: 31804482 PMCID: PMC6895051 DOI: 10.1038/s41467-019-13485-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 11/08/2019] [Indexed: 02/08/2023] Open
Abstract
Overexpressed Aurora-A kinase promotes tumor growth through various pathways, but whether Aurora-A is also involved in metabolic reprogramming-mediated cancer progression remains unknown. Here, we report that Aurora-A directly interacts with and phosphorylates lactate dehydrogenase B (LDHB), a subunit of the tetrameric enzyme LDH that catalyzes the interconversion between pyruvate and lactate. Aurora-A-mediated phosphorylation of LDHB serine 162 significantly increases its activity in reducing pyruvate to lactate, which efficiently promotes NAD+ regeneration, glycolytic flux, lactate production and bio-synthesis with glycolytic intermediates. Mechanistically, LDHB serine 162 phosphorylation relieves its substrate inhibition effect by pyruvate, resulting in remarkable elevation in the conversions of pyruvate and NADH to lactate and NAD+. Blocking S162 phosphorylation by expression of a LDHB-S162A mutant inhibited glycolysis and tumor growth in cancer cells and xenograft models. This study uncovers a function of Aurora-A in glycolytic modulation and a mechanism through which LDHB directly contributes to the Warburg effect. Aurora-A kinase is frequently over-expressed in tumours. Here, the authors show that it modulates the activity of lactate dehydrogenase B, resulting in enhanced glycolysis, bio-synthesis and tumour growth.
Collapse
Affiliation(s)
- Aoxing Cheng
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Peng Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Dongdong Yang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaotao Duan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yongliang Jiang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tian Xu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ya Jiang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiahui Shi
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chengtao Ding
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Gao Wu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhihong Sang
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Qiang Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mian Wu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhiyong Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin Pan
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Yue-Yin Pan
- Department of Medical Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ping Gao
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huafeng Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cong-Zhao Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Guo
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Zhenye Yang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS key Laboratory of Innate Immunity and Chronic Disease, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
20
|
Huang Y, Ha S, Li Z, Li J, Xiao W. CHK1-CENP B/MAD2 is associated with mild oxidative damage-induced sex chromosome aneuploidy of male mouse embryos during in vitro fertilization. Free Radic Biol Med 2019; 137:181-193. [PMID: 31042615 DOI: 10.1016/j.freeradbiomed.2019.04.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 02/05/2023]
Abstract
A high incidence of aneuploidy is observed in vitro fertilization (IVF)-derived embryos, but the formation and repair mechanisms are unknown. Here, we investigated the effects of slightly increased reactive oxygen species (ROS) produced by in vitro culture conditions on embryo aneuploidy and the roles of the spindle assembly checkpoint (SAC) protein, mitotic arrest-deficient 2 (MAD2), and the DNA damage response (DDR) protein, checkpoint kinase 1 (CHK1), in aneuploidy repair. By assessing chromosome abnormalities via DAPI staining, karyotype analysis and next-generation sequencing technology, we demonstrated that mild oxidative damage mainly increased the risk of sex chromosome aneuploidy in male mouse embryos (41,XXY,+X and 41,XYY,+Y) through chromosome mis-segregation during the first mitosis. Isobaric tags for relative and absolute quantitation technology revealed that mild oxidative damage inhibited the expression of male reproduction-related proteins, including a kinase anchor protein 4 (AKAP4), whose gene is located on mouse/human Chromosome X. Under mild oxidative damage, abrogation of MAD2 by MAD2 inhibitor-1 (M2I-1) or CHK1 by siRNA microinjection increased sex chromosome mosaicism rate and reduced mitosis-promoting factor (MPF) activity. CHK1 inhibition also reduced kinetochore localization of centromere protein B (CENP B) and MAD2. These findings show that DDR and SAC are responsible for repair of sex chromosome mosaicism via the pCHK1 (S345)-CENP B/MAD2-MPF pathway; further, IVF may have negative effects on male offspring's reproduction ability, which ultimately depends on their continued repair capability. Therefore, we suggest that antioxidants, especially those targeting improved CHK1-MAD2 function, may be a promising therapeutic strategy to reduce aneuploidy formation of IVF-derived embryos and to maintain genome integrity of embryo and offspring.
Collapse
Affiliation(s)
- Yue Huang
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, 515000, Guangdong, China
| | - Siyao Ha
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics & Gynecology, Fudan University Shanghai Medical College, Shanghai, 200011, China
| | - Zhiling Li
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, 515000, Guangdong, China.
| | - Jiena Li
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, 515000, Guangdong, China
| | - Wanfen Xiao
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, 515000, Guangdong, China
| |
Collapse
|
21
|
Magnaghi-Jaulin L, Eot-Houllier G, Gallaud E, Giet R. Aurora A Protein Kinase: To the Centrosome and Beyond. Biomolecules 2019; 9:biom9010028. [PMID: 30650622 PMCID: PMC6359016 DOI: 10.3390/biom9010028] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Accurate chromosome segregation requires the perfect spatiotemporal rearrangement of the cellular cytoskeleton. Isolated more than two decades ago from Drosophila, Aurora A is a widespread protein kinase that plays key roles during cell division. Numerous studies have described the localisation of Aurora A at centrosomes, the mitotic spindle, and, more recently, at mitotic centromeres. In this review, we will summarise the cytoskeletal rearrangements regulated by Aurora A during cell division. We will also discuss the recent discoveries showing that Aurora A also controls not only the dynamics of the cortical proteins but also regulates the centromeric proteins, revealing new roles for this kinase during cell division.
Collapse
Affiliation(s)
- Laura Magnaghi-Jaulin
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Grégory Eot-Houllier
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Emmanuel Gallaud
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Régis Giet
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| |
Collapse
|
22
|
Su CC, Chen NC, Chyau CC, Tseng HC, Chou FP. Induction of Mitotic Catastrophe via Inhibition of Aurora B by Ionizing Radiation With Additive of Mulberry Water Extract in Human Bladder Cancer Cells. Integr Cancer Ther 2018; 18:1534735418808586. [PMID: 30428726 PMCID: PMC7290068 DOI: 10.1177/1534735418808586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mulberry fruit water extract (MWE) has been reported to synergistically enhance the cytotoxic effect of paclitaxel by promoting mitotic catastrophe to induce apoptosis in bladder cancer cells in our previous work. The aim of this study was to evaluate and to mechanistically explore the effects of MWE on bladder cancer responses to ionizing radiation (IR) by treating TSGH 8301 bladder carcinoma cells with MWE after exposing to IR. The results of MTT assay showed a synergistic cytotoxicity of IR with the co-treatment of MWE (IR/MWE) by inducing G2/M phase arrest as demonstrated by flow cytometry analysis in TSGH 8301, HT1367 and HT1197 bladder carcinoma cells lines. The IR/MWE-treated cells expressed increased levels of the G2/M phase arrest-related proteins cdc2/cyclin B1 and displayed giant multinucleated morphology, a typical characteristic of mitotic catastrophe. Immunofluorescent confocal microscopy revealed that the combined strategy inhibited Aurora B phosphorylation through Ras/Raf/MEK/ERK signaling cascade as demonstrated by Western blotting analysis. IR/MWE also caused an inhibitory effect on Plk1 and the subsequent downstream regulator RhoA repression and Cep55 induction, which would influence cell cycle progression in the early steps of cytokinesis. A profound tumor growth suppression and inactivation of Aurora B activity in the tumor tissues by IR/MWE treatment were confirmed in the TSGH 8301 xenograft model in vivo. These data demonstrated that MWE could be an effective auxiliary to synergize with radiation on the anticancer efficacy by promoting mitotic catastrophe through inhibition of Aurora B, providing a novel and effective therapeutic option for bladder cancer management.
Collapse
Affiliation(s)
- Chia-Cheng Su
- 1 Chi Mei Medical Center, Tainan City, Taiwan.,2 Chia Nan University of Pharmacy & Science, Tainan City, Taiwan
| | | | | | - Hsien-Chun Tseng
- 3 Chung Shan Medical University, Taichung City, Taiwan.,5 Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Fen-Pi Chou
- 3 Chung Shan Medical University, Taichung City, Taiwan.,5 Chung Shan Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|
23
|
Pyrazolo[4,3-b]pyrimido[4,5-e][1,4]diazepine derivatives as new multi-targeted inhibitors of Aurora A/B and KDR. Eur J Med Chem 2018; 158:428-441. [DOI: 10.1016/j.ejmech.2018.09.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 08/16/2018] [Accepted: 09/10/2018] [Indexed: 12/19/2022]
|
24
|
Courtheoux T, Diallo A, Damodaran AP, Reboutier D, Watrin E, Prigent C. Aurora A kinase activity is required to maintain an active spindle assembly checkpoint during prometaphase. J Cell Sci 2018; 131:jcs.191353. [PMID: 29555820 DOI: 10.1242/jcs.191353] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
During the prometaphase stage of mitosis, the cell builds a bipolar spindle of microtubules that mechanically segregates sister chromatids between two daughter cells in anaphase. The spindle assembly checkpoint (SAC) is a quality control mechanism that monitors proper attachment of microtubules to chromosome kinetochores during prometaphase. Segregation occurs only when each chromosome is bi-oriented with each kinetochore pair attached to microtubules emanating from opposite spindle poles. Overexpression of the protein kinase Aurora A is a feature of various cancers and is thought to enable tumour cells to bypass the SAC, leading to aneuploidy. Here, we took advantage of a chemical and chemical-genetic approach to specifically inhibit Aurora A kinase activity in late prometaphase. We observed that a loss of Aurora A activity directly affects SAC function, that Aurora A is essential for maintaining the checkpoint protein Mad2 on unattached kinetochores and that inhibition of Aurora A leads to loss of the SAC, even in the presence of nocodazole or Taxol. This is a new finding that should affect the way Aurora A inhibitors are used in cancer treatments.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Thibault Courtheoux
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Alghassimou Diallo
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Arun Prasath Damodaran
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - David Reboutier
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Erwan Watrin
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Claude Prigent
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| |
Collapse
|
25
|
Lane SIR, Jones KT. Chromosome biorientation and APC activity remain uncoupled in oocytes with reduced volume. J Cell Biol 2017; 216:3949-3957. [PMID: 28978643 PMCID: PMC5716262 DOI: 10.1083/jcb.201606134] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/28/2017] [Accepted: 09/07/2017] [Indexed: 11/23/2022] Open
Abstract
Lane and Jones use serial bisection of mouse oocytes to analyze the influence of cytoplasmic volume on spindle assembly checkpoint function. Volume reduction promotes inhibition of APC but cannot prevent chromosome segregation errors at anaphase. The spindle assembly checkpoint (SAC) prevents chromosome missegregation by coupling anaphase onset with correct chromosome attachment and tension to microtubules. It does this by generating a diffusible signal from free kinetochores into the cytoplasm, inhibiting the anaphase-promoting complex (APC). The volume in which this signal remains effective is unknown. This raises the possibility that cell volume may be the reason the SAC is weak, and chromosome segregation error-prone, in mammalian oocytes. Here, by a process of serial bisection, we analyzed the influence of oocyte volume on the ability of the SAC to inhibit bivalent segregation in meiosis I. We were able to generate oocytes with cytoplasmic volumes reduced by 86% and observed changes in APC activity consistent with increased SAC control. However, bivalent biorientation remained uncoupled from APC activity, leading to error-prone chromosome segregation. We conclude that volume is one factor contributing to SAC weakness in oocytes. However, additional factors likely uncouple chromosome biorientation with APC activity.
Collapse
Affiliation(s)
- Simon I R Lane
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK .,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Keith T Jones
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|