1
|
Lin J, Zhu Y, Lin Z, Yu J, Lin X, Lai W, Tong B, Xu L, Li E, Long L. The Expression Regulation and Cancer-Promoting Roles of RACGAP1. Biomolecules 2024; 15:3. [PMID: 39858398 PMCID: PMC11760467 DOI: 10.3390/biom15010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 01/27/2025] Open
Abstract
RACGAP1 is a Rho-GTPase-activating protein originally discovered in male germ cells to inactivate Rac, RhoA and Cdc42 from the GTP-bound form to the GDP-bound form. GAP has traditionally been known as a tumor suppressor. However, studies increasingly suggest that overexpressed RACGAP1 activates Rac and RhoA in multiple cancers to mediate downstream oncogene overexpression by assisting in the nuclear translocation of signaling molecules and to promote cytokinesis by regulating the cytoskeleton or serving as a component of the central spindle. Contradictorily, it was also reported that RACGAP1 in gastric cancer could inactivate Rac and RhoA. In addition, studies have revealed that RACGAP1 can be a biomarker for prognosis, and its role in reducing doxorubicin sensitivity poses difficulties for treatment, while the current drug targets mainly focus on its downstream molecule. This article mainly reviews the expression regulation of RACGAP1 and its cancer-promoting functions through oncogene expression mediation and Rho-GTPase activation.
Collapse
Affiliation(s)
- Jiacheng Lin
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Yuhao Zhu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Zhaoping Lin
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Jindong Yu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Xiaobing Lin
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Weiyuan Lai
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Beibei Tong
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Liyan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Lin Long
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
2
|
Ma N, Wang H, Lu Q, Liu J, Fan X, Li L, Wang Q, Li X, Yu B, Zhang Y, Gao J. Temporal changes of neurobehavior in rats following varied blast magnitudes and screening of serum biomarkers in early stage of brain injury. Sci Rep 2024; 14:30023. [PMID: 39627295 PMCID: PMC11615197 DOI: 10.1038/s41598-024-81656-9] [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: 08/27/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024] Open
Abstract
Blast neurotrauma has been linked to impairments in higher-order cognitive functions, including memory, attention, and mood. Current literature is limited to a single overpressure exposure or repeated exposures at the same level of overpressure. In this study, a rodent model of primary blast neurotrauma was employed to determine the pressure at which acute and chronic neurological alterations occurred. Three pressure magnitudes (low, moderate and high) were used to evaluate injury thresholds. A biology shock tube (BST) was used to simulate shock waves with overpressures of 60 kPa, 90 kPa and 120 kPa respectively. Neurological behavior of the rats was assessed by the Multi-Conditioning System (MCS) at 1 d, 7 d, 28 d and 90 d after shock wave exposure. Serum dopamine (DA), 5-hydroxytryptamine (5-HT), brain-derived neurotrophic factor (BDNF) and gamma-aminobutyric acid (GABA) were measured at the same time points. The proteomic analysis was conducted to identify potentially vulnerable cellular and molecule targets of serum in the immediate post-exposure period. Results revealed that: (1) Anxiety-like behavior increased significantly at 1 d post-exposure in the medium and high overpressure (90 kPa, 120 kPa) groups, returned to baseline at 7 days, and anxiety-like behavior in the high overpressure groups re-emerged at 28 d and 90 d. (2) High overpressure (120 kPa) impaired learning and memory in the immediate post-exposure period. (3) The serum DA levels decreased significantly at 1 d post-exposure in the medium and high overpressure groups; The 5-HT levels decreased significantly at 1 d and 90 d in the high overpressure groups; The BDNF levels decreased significantly at 90 d in the high overpressure groups. (4) Proteomic analysis identified 38, 306, and 57 differentially expressed proteins in serum following low, medium and high overpressure exposures, respectively. Two co-expressed proteins were validated. Functional analysis revealed significant enrichment of 1121, 2096, and 1121 Gene Ontology (GO) items and 33, 47, and 26 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, indicating extensive molecular responses to overpressure in the early phase. These findings suggest that exposure, even at moderate levels, can induce persistent neurobehavioral and molecular alterations, highlighting the need for further research into the long-term consequences of blast neurotrauma.
Collapse
Affiliation(s)
- Ning Ma
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Hong Wang
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Qing Lu
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Jinren Liu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Xiaolin Fan
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Liang Li
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Qi Wang
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Xiao Li
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Boya Yu
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Yuhao Zhang
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China
| | - Junhong Gao
- Xi'an Key Laboratory of Toxicology and Biological Effects, Research Center for Toxicological and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, 710065, China.
| |
Collapse
|
3
|
Wang T, Wang S, Li Z, Xie J, Chen H, Hou J. Machine learning-informed liquid-liquid phase separation for personalized breast cancer treatment assessment. Front Immunol 2024; 15:1485123. [PMID: 39628476 PMCID: PMC11611825 DOI: 10.3389/fimmu.2024.1485123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
Background Breast cancer, characterized by its heterogeneity, is a leading cause of mortality among women. The study aims to develop a Machine Learning-Derived Liquid-Liquid Phase Separation (MDLS) model to enhance the prognostic accuracy and personalized treatment strategies for breast cancer patients. Methods The study employed ten machine learning algorithms to construct 108 algorithm combinations for the MDLS model. The robustness of the model was evaluated using multi-omics and single-cell data across 14 breast cancer cohorts, involving 9,723 patients. Genetic mutation, copy number alterations, and single-cell RNA sequencing were analyzed to understand the molecular mechanisms and predictive capabilities of the MDLS model. Immunotherapy targets were predicted by evaluating immune cell infiltration and immune checkpoint expression. Chemotherapy targets were identified through correlation analysis and drug responsiveness prediction. Results The MDLS model demonstrated superior prognostic power, with a mean C-index of 0.649, outperforming 69 published signatures across ten cohorts. High-MDLS patients exhibited higher tumor mutation burden and distinct genomic alterations, including significant gene amplifications and deletions. Single-cell analysis revealed higher MDLS activity in tumor-aneuploid cells and identified key regulatory factors involved in MDLS progression. Cell-cell communication analysis indicated stronger interactions in high-MDLS groups, and immunotherapy response evaluation showed better outcomes for low-MDLS patients. Conclusion The MDLS model offers a robust and precise tool for predicting breast cancer prognosis and tailoring personalized treatment strategies. Its integration of multi-omics and machine learning highlights its potential clinical applications, particularly in improving the effectiveness of immunotherapy and identifying therapeutic targets for high-MDLS patients.
Collapse
Affiliation(s)
- Tao Wang
- Research Laboratory Center, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Shu Wang
- Department of Breast Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zhuolin Li
- Department of Breast Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jie Xie
- Department of Breast Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Huan Chen
- Department of Breast Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jing Hou
- Department of Breast Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
4
|
Xu Q, Li J, Zhuo L, Gao H, Yang Y, Li W. RACGAP1 is a pivotal gene in lung adenocarcinoma-associated membranous nephropathy: Based on comprehensive bioinformatics analysis and machine learning. Int Immunopharmacol 2024; 139:112783. [PMID: 39068752 DOI: 10.1016/j.intimp.2024.112783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND This study performs a detailed bioinformatics and machine learning analysis to investigate the genetic foundations of membranous nephropathy (MN) in lung adenocarcinoma (LUAD). METHODS In this study, the gene expression profiles of MN microarray datasets (GSE99339) and LUAD dataset (GSE43767) were downloaded from the Gene Expression Omnibus database, common differentially expressed genes (DEGs) were obtained using the limma R package. The biological functions were analyzed with R Cluster Profiler package according to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Machine learning algorithms, including LASSO regression, support vector machine (SVM), Random Forest, and Boruta analysis, were applied to identify hubgenes linked to LUAD-associated MN. These genes' prognostic values were evaluated in the TCGA-LUAD cohort and validated through immunohistochemistry on renal biopsy specimens. RESULTS A total of 36 DEGs in common were identified for downstream analyses. Functional enrichment analysis highlighted the involvement of the Toll-like receptor 4 pathway and several immune recognition pathways in LUAD-associated MN. COL3A1, PSENEN, RACGAP1, and TNFRSF10B were identified as hub genes in LUAD-associated MN using machine learning algorithms. ROC analysis demonstrated their effective discrimination of MN with high accuracy. Survival analysis showed that lung adenocarcinoma patients with higher expression of these genes had significantly reduced overall survival. In patients with lung adenocarcinoma-associated MN, RACGAP1, COL3A1, PSENEN, and TNFRSF10B were higher expressed in the glomerular, especially RACGAP1, indicating an important role in the pathogenesis of LUAD-associated membranous nephropathy. CONCLUSIONS Our study underscores the critical role of RACGAP1, COL3A1, PSENEN, and TNFRSF10B in the development of LUAD-associated MN, providing important insights for future research and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Qianqian Xu
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Jiayi Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China; Department of Nephrology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Li Zhuo
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Hongmei Gao
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Yue Yang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China; Department of Nephrology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| |
Collapse
|
5
|
Wang J, Liu H, Yu Z, Zhou Q, Sun F, Han J, Gao L, Dou B, Zhang H, Fu J, Jia W, Chen W, Hu J, Han B. Reciprocal regulation between RACGAP1 and AR contributes to endocrine therapy resistance in prostate cancer. Cell Commun Signal 2024; 22:339. [PMID: 38898473 PMCID: PMC11186203 DOI: 10.1186/s12964-024-01703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Endocrine resistance driven by sustained activation of androgen receptor (AR) signaling pathway in advanced prostate cancer (PCa) is fatal. Characterization of mechanisms underlying aberrant AR pathway activation to search for potential therapeutic strategy is particularly important. Rac GTPase-activating protein 1 (RACGAP1) is one of the specific GTPase-activating proteins. As a novel tumor proto-oncogene, overexpression of RACGAP1 was related to the occurrence of various tumors. METHODS Bioinformatics methods were used to analyze the relationship of expression level between RACGAP1 and AR as well as AR pathway activation. qRT-PCR and western blotting assays were performed to assess the expression of AR/AR-V7 and RACGAP1 in PCa cells. Immunoprecipitation and immunofluorescence experiments were conducted to detect the interaction and co-localization between RACGAP1 and AR/AR-V7. Gain- and loss-of-function analyses were conducted to investigate the biological roles of RACGAP1 in PCa cells, using MTS and colony formation assays. In vivo experiments were conducted to evaluate the effect of RACGAP1 inhibition on the tumor growth. RESULTS RACGAP1 was a gene activated by AR, which was markedly upregulated in PCa patients with CRPC and enzalutamide resistance. AR transcriptionally activated RACGAP1 expression by binding to its promoter region. Reciprocally, nuclear RACGAP1 bound to the N-terminal domain (NTD) of both AR and AR-V7, blocking their interaction with the E3 ubiquitin ligase MDM2. Consequently, this prevented the degradation of AR/AR-V7 in a ubiquitin-proteasome-dependent pathway. Notably, the positive feedback loop between RACGAP1 and AR/AR-V7 contributed to endocrine therapy resistance of CRPC. Combination of enzalutamide and in vivo cholesterol-conjugated RIG-I siRNA drugs targeting RACGAP1 induced potent inhibition of xenograft tumor growth of PCa. CONCLUSION In summary, our results reveal that reciprocal regulation between RACGAP1 and AR/AR-V7 contributes to the endocrine resistance in PCa. These findings highlight the therapeutic potential of combined RACGAP1 inhibition and enzalutamide in treatment of advanced PCa.
Collapse
Affiliation(s)
- Jiajia Wang
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Hui Liu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Zeyuan Yu
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Qianqian Zhou
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Feifei Sun
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Jingying Han
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Lin Gao
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Baokai Dou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hanwen Zhang
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Jiawei Fu
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
| | - Wenqiao Jia
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Hu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China.
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Department of Pathology, School of Basic Medical Sciences, Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
6
|
Xu Z, Wu S, Tu J, Wang M, Liang W, Cheng J, Guan J, Xu J. RACGAP1 promotes lung cancer cell proliferation through the PI3K/AKT signaling pathway. Sci Rep 2024; 14:8694. [PMID: 38622149 PMCID: PMC11018837 DOI: 10.1038/s41598-024-58539-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
We aimed to investigate the expression and clinic significance of Rac GTPase Activating Protein 1 (RACGAP1) in human lung adenocarcinoma (LUAD). Online database analysis revealed a significant increase in RACGAP1 mRNA expression among 26 types of tumor tissues, including LUAD tissues. Online database and tissue microarray analyses indicated that RACGAP1 expression was significantly upregulated in LUAD tissues. Genetic variation analysis identified four different genetic variations of RACGAPs in LUAD. Moreover, online database analysis showed that RACGAP1 upregulation was correlated with shorter survival in patients with LUAD. After silencing RACGAP1 expression in A549 cells using siRNA and assessing its protein levels via Western blotting, we found that RACGAP1 knockdown inhibited cell growth and induced apoptosis determined using the Cell Counting Kit-8 assay, colony formation assay, and flow cytometry. Mechanistically, western blot analysis indicated that Bax expression increased, whereas Bcl-2 expression decreased. Moreover, RACGAP1 knockdown attenuated PI3K/AKT pathway activation in lung cancer cells. Taken together, our findings showed that RACGAP1 was overexpressed in LUAD tissues and played an important role in lung cancer by increasing cell growth through the PI3K/AKT signaling pathway. This study suggests recommends evaluating RACGAP1 in clinical settings as a novel biomarker and potential therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Zhiyang Xu
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China
| | - Shaohang Wu
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China
| | - Jiahua Tu
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China
| | - Mingyang Wang
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China
| | - Weicheng Liang
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China
| | - Jiangdong Cheng
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China
| | - Jun Guan
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China.
| | - Jianxin Xu
- Department of Thoracic Surgery, The First Hospital of Putian, The School of Clinical Medicine, Fujian Medical University Putian, Fujian, 351100, China.
| |
Collapse
|
7
|
Yuan Q, Li L, Wang LS, Xing SG. Epidemiological and transcriptome data identify shared gene signatures and immune cell infiltration in type 2 diabetes and non-small cell lung cancer. Diabetol Metab Syndr 2024; 16:64. [PMID: 38468345 DOI: 10.1186/s13098-024-01278-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/25/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Numerous previous studies have reported an association between type 2 diabetes mellitus (T2DM) and lung cancer risk, but the underlying mechanism of the interaction remains unclear. This study aimed to investigate the shared genetic features and immune infiltration processes between lung cancer and T2DM. METHODS Epidemiological data from the National Health and Nutrition Examination Survey (NHANES) 2000-2018 was used to explore the relationship between lung cancer and diabetes systematically. In addition, we also used bioinformatics methods to analyze the transcriptome data from the Gene Expression Omnibus (GEO) to explore the potential functional mechanisms from the perspective of genes and immune infiltration. RESULTS Logistic regression analysis showed that prediabetes (OR = 3.289,95%CI 1.231, 8.788, p = 0.01760, model 3)and type 2 diabetes (OR = 3.032 95%CI,1.015, 9.054, p = 0.04689) were significantly associated with an increased risk of lung cancer after adjusting for multiple covariates. Data from NHANES showed an inverted U-shaped relationship between fasting blood glucose and glycosylated haemoglobin and the risk of lung cancer (P for non-linear < 0.001). Transcriptome data showed that we screened 57 co-DEGs, of which 25 were up-regulated co-DEGs and 32 were down-regulated. Ten core DEGs were identified by bioinformatics analysis, which were SMC6, CDC27, CDC7, RACGAP1, SMC4, NCF4, NCF1, NCF2, SELPLG and CFP. Correlation analysis showed that some core DEGs were significantly associated with simultaneous dysregulation of immune cells. CONCLUSION The identified core genes of NSCLC and T2DM are associated with dysregulated immune cells, which provides a potential research avenue for diagnosing and treating lung cancer combined with diabetes.
Collapse
Affiliation(s)
- Qian Yuan
- Department of Thoracic surgery, Nan Jing Gaochun people's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), 210000, Nanjing, Jiangsu, China
| | - Long Li
- Department of Thoracic surgery, Nan Jing Gaochun people's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), 210000, Nanjing, Jiangsu, China
| | - Liu-Shun Wang
- Department of Thoracic surgery, Nan Jing Gaochun people's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), 210000, Nanjing, Jiangsu, China
| | - Shi-Gui Xing
- Department of Thoracic surgery, Nan Jing Gaochun people's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), 210000, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Warecki B, Tao L. Centralspindlin-mediated transport of RhoGEF positions the cleavage plane for cytokinesis. Sci Signal 2023; 16:eadh0601. [PMID: 37402224 PMCID: PMC10501416 DOI: 10.1126/scisignal.adh0601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/13/2023] [Indexed: 07/06/2023]
Abstract
During cytokinesis, the cell membrane furrows inward along a cleavage plane. The positioning of the cleavage plane is critical to faithful cell division and is determined by the Rho guanine nucleotide exchange factor (RhoGEF)-mediated activation of the small guanosine triphosphatase RhoA and the conserved motor protein complex centralspindlin. Here, we explored whether and how centralspindlin mediates the positioning of RhoGEF. In dividing neuroblasts from Drosophila melanogaster, we observed that immediately before cleavage, first centralspindlin and then RhoGEF localized to the sites where cleavage subsequently initiated. Using in vitro assays with purified Drosophila proteins and stabilized microtubules, we found that centralspindlin directly transported RhoGEF as cargo along single microtubules and sequestered it at microtubule plus-ends for prolonged periods of time. In addition, the binding of RhoGEF to centralspindlin appeared to stimulate centralspindlin motor activity. Thus, the motor activity and microtubule association of centralspindlin can translocate RhoGEF to areas where microtubule plus-ends are abundant, such as at overlapping astral microtubules, to locally activate RhoA and accurately position the cleavage plane during cell division.
Collapse
Affiliation(s)
- Brandt Warecki
- Department of Biology, University of Hawai’i at Hilo, HI 96720, USA
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz; Santa Cruz, CA 95064, USA
| | - Li Tao
- Department of Biology, University of Hawai’i at Hilo, HI 96720, USA
| |
Collapse
|
9
|
Bryl R, Nawrocki MJ, Jopek K, Kaczmarek M, Bukowska D, Antosik P, Mozdziak P, Zabel M, Dzięgiel P, Kempisty B. Transcriptomic Characterization of Genes Regulating the Stemness in Porcine Atrial Cardiomyocytes during Primary In Vitro Culture. Genes (Basel) 2023; 14:1223. [PMID: 37372403 PMCID: PMC10297922 DOI: 10.3390/genes14061223] [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: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Heart failure remains a major cause of death worldwide. There is a need to establish new management options as current treatment is frequently suboptimal. Clinical approaches based on autologous stem cell transplant is potentially a good alternative. The heart was long considered an organ unable to regenerate and renew. However, several reports imply that it may possess modest intrinsic regenerative potential. To allow for detailed characterization of cell cultures, whole transcriptome profiling was performed after 0, 7, 15, and 30 days of in vitro cell cultures (IVC) from the right atrial appendage and right atrial wall utilizing microarray technology. In total, 4239 differentially expressed genes (DEGs) with ratio > abs |2| and adjusted p-value ≤ 0.05 for the right atrial wall and 4662 DEGs for the right atrial appendage were identified. It was shown that a subset of DEGs, which have demonstrated some regulation of expression levels with the duration of the cell culture, were enriched in the following GO BP (Gene Ontology Biological Process) terms: "stem cell population maintenance" and "stem cell proliferation". The results were validated by RT-qPCR. The establishment and detailed characterization of in vitro culture of myocardial cells may be important for future applications of these cells in heart regeneration processes.
Collapse
Affiliation(s)
- Rut Bryl
- Section of Regenerative Medicine and Cancer Research, Natural Sciences Club, Faculty of Biology, Adam Mickiewicz University, Poznań, 61-614 Poznan, Poland;
| | - Mariusz J. Nawrocki
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland;
- Gene Therapy Laboratory, Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
| | - Maciej Zabel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.Z.); (P.D.)
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Piotr Dzięgiel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.Z.); (P.D.)
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Obstetrics and Gynaecology, University Hospital and Masaryk University, 62500 Brno, Czech Republic
| |
Collapse
|
10
|
di Pietro F, Osswald M, De Las Heras JM, Cristo I, López-Gay J, Wang Z, Pelletier S, Gaugué I, Leroy A, Martin C, Morais-de-Sá E, Bellaïche Y. Systematic analysis of RhoGEF/GAP localizations uncovers regulators of mechanosensing and junction formation during epithelial cell division. Curr Biol 2023; 33:858-874.e7. [PMID: 36917931 PMCID: PMC10017266 DOI: 10.1016/j.cub.2023.01.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/30/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023]
Abstract
Cell proliferation is central to epithelial tissue development, repair, and homeostasis. During cell division, small RhoGTPases control both actomyosin dynamics and cell-cell junction remodeling to faithfully segregate the genome while maintaining tissue polarity and integrity. To decipher the mechanisms of RhoGTPase spatiotemporal regulation during epithelial cell division, we generated a transgenic fluorescently tagged library for the 48 Drosophila Rho guanine exchange factors (RhoGEFs) and GTPase-activating proteins (GAPs), and we systematically characterized their endogenous distributions by time-lapse microscopy. Therefore, we unveiled candidate regulators of the interplay between actomyosin and junctional dynamics during epithelial cell division. Building on these findings, we established that the conserved RhoGEF Cysts and RhoGEF4 play sequential and distinct roles to couple cytokinesis with de novo junction formation. During ring contraction, Cysts via Rho1 participates in the neighbor mechanosensing response, promoting daughter-daughter cell membrane juxtaposition in preparation to de novo junction formation. Subsequently and upon midbody formation, RhoGEF4 via Rac acts in the dividing cell to ensure the withdrawal of the neighboring cell membranes, thus controlling de novo junction length and cell-cell arrangements upon cytokinesis. Altogether, our findings delineate how the RhoGTPases Rho and Rac are locally and temporally activated during epithelial cytokinesis, highlighting the RhoGEF/GAP library as a key resource to understand the broad range of biological processes regulated by RhoGTPases.
Collapse
Affiliation(s)
- Florencia di Pietro
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Mariana Osswald
- IBMC - Instituto de Biologia Molecular e Celular; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - José M De Las Heras
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Inês Cristo
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Jesús López-Gay
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Zhimin Wang
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Stéphane Pelletier
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Isabelle Gaugué
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Adrien Leroy
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Charlotte Martin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Eurico Morais-de-Sá
- IBMC - Instituto de Biologia Molecular e Celular; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Yohanns Bellaïche
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France.
| |
Collapse
|
11
|
Yang C, Chen L, Niu Q, Ge Q, Zhang J, Tao J, Zhou J, Liang C. Identification and validation of an E2F-related gene signature for predicting recurrence-free survival in human prostate cancer. Cancer Cell Int 2022; 22:382. [PMID: 36471446 PMCID: PMC9721026 DOI: 10.1186/s12935-022-02791-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND It is well-established that biochemical recurrence is detrimental to prostate cancer (PCa). In the present study, we explored the mechanisms underlying PCa progression. METHODS Five cohorts from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases were used to perform gene set variation analysis (GSVA) between nonrecurrent and recurrent PCa patients. We obtained the intersection of pathway enrichment results and extracted the corresponding gene list. LASSO Cox regression analysis was used to identify recurrence-free survival (RFS)-related significant genes and establish an RFS prediction gene signature and nomogram. MTT and colony formation assays were conducted to validate our findings. RESULTS The E2F signaling pathway was activated in recurrent PCa patients compared to nonrecurrent patients. We established an E2F-related gene signature for RFS prediction based on the four identified E2F-related genes (CDKN2C, CDKN3, RACGAP1, and RRM2) using LASSO Cox regression in the Memorial Sloan Kettering Cancer Center (MSKCC) cohort. The risk score of each patient in MSKCC was calculated based on the expression levels of CDKN2C, CDKN3, RACGAP1, and RRM2. PCa patients with low-risk scores exhibited higher RFS than those with high-risk scores. Receiver operating characteristic (ROC) curve analysis validated the good performance and prognostic accuracy of the E2F-related gene signature, which was validated in the TCGA-prostate adenocarcinoma (TCGA-PRAD) cohort. Compared to patients with low Gleason scores and early T stages, PCa patients with high Gleason scores and advanced T stages had high-risk scores. Moreover, the E2F-related gene signature-based nomogram yielded good performance in RFS prediction. Functional experiments further confirmed these results. CONCLUSIONS The E2F signaling pathway is associated with biochemical recurrence in PCa. Our established E2F-related gene signature and nomogram yielded good accuracy in predicting the biochemical recurrence in PCa.
Collapse
Affiliation(s)
- Cheng Yang
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| | - Lei Chen
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| | - Qingsong Niu
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| | - Qintao Ge
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| | - Jiong Zhang
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| | - Junyue Tao
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| | - Jun Zhou
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| | - Chaozhao Liang
- grid.412679.f0000 0004 1771 3402Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XInstitute of Urology, Anhui Medical University, Hefei, China ,grid.186775.a0000 0000 9490 772XAnhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218, Shushan District, Hefei City, 230022 Anhui Province People’s Republic of China
| |
Collapse
|
12
|
Minor Kinases with Major Roles in Cytokinesis Regulation. Cells 2022; 11:cells11223639. [PMID: 36429067 PMCID: PMC9688779 DOI: 10.3390/cells11223639] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Cytokinesis, the conclusive act of cell division, allows cytoplasmic organelles and chromosomes to be faithfully partitioned between two daughter cells. In animal organisms, its accurate regulation is a fundamental task for normal development and for preventing aneuploidy. Cytokinesis failures produce genetically unstable tetraploid cells and ultimately result in chromosome instability, a hallmark of cancer cells. In animal cells, the assembly and constriction of an actomyosin ring drive cleavage furrow ingression, resulting in the formation of a cytoplasmic intercellular bridge, which is severed during abscission, the final event of cytokinesis. Kinase-mediated phosphorylation is a crucial process to orchestrate the spatio-temporal regulation of the different stages of cytokinesis. Several kinases have been described in the literature, such as cyclin-dependent kinase, polo-like kinase 1, and Aurora B, regulating both furrow ingression and/or abscission. However, others exist, with well-established roles in cell-cycle progression but whose specific role in cytokinesis has been poorly investigated, leading to considering these kinases as "minor" actors in this process. Yet, they deserve additional attention, as they might disclose unexpected routes of cell division regulation. Here, we summarize the role of multifunctional kinases in cytokinesis with a special focus on those with a still scarcely defined function during cell cleavage. Moreover, we discuss their implication in cancer.
Collapse
|
13
|
Eid RA, Soltan MA, Eldeen MA, Shati AA, Dawood SA, Eissa M, Zaki MSA, Algahtani M, Theyab A, Abdel-Daim MM, Kim B. Assessment of RACGAP1 as a Prognostic and Immunological Biomarker in Multiple Human Tumors: A Multiomics Analysis. Int J Mol Sci 2022; 23:ijms232214102. [PMID: 36430577 PMCID: PMC9695706 DOI: 10.3390/ijms232214102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
Several recent studies have pointed out that arc GTPase activating protein 1 (RACGAP1) is a putative oncogene in many human tumors. However, to date, no pan-cancer analysis has been performed to study the different aspects of this gene expression and behavior in tumor tissues. Here, we applied several bioinformatics tools to perform a comprehensive analysis for RACGAP1. First, we assessed the expression of RACGAP1 in several types of human tumors and tried to correlate that with the stage of the tumors analyzed. We then performed a survival analysis to study the correlation between RACGAP1 upregulation in tumors and the clinical outcome. Additionally, we investigated the mutation forms, the correlation with several immune cell infiltration, the phosphorylation status of the interested protein in normal and tumor tissues, and the potential molecular mechanisms of RACGAP1 in cancerous tissue. The results demonstrated that RACGAP1, a highly expressed gene across several types of tumors, correlated with a poor prognosis in several types of human cancers. Moreover, it was found that RACGAP1 affects the tumor immune microenvironment by influencing the infiltration level of several immune cells. Collectively, the current study provides a comprehensive overview of the oncogenic roles of RACGAP1, where our results nominate it as a potential prognostic biomarker and a target for antitumor therapy development.
Collapse
Affiliation(s)
- Refaat A. Eid
- Pathology Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
| | - Mohamed A. Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia 41611, Egypt
| | - Muhammad Alaa Eldeen
- Cell Biology, Histology & Genetics Division, Biology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
- Correspondence: (M.A.E.); (B.K.)
| | - Ayed A. Shati
- Department of Child Health, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
| | - Samy A. Dawood
- Department of Child Health, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
| | - Mohamed Eissa
- Pathology Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed Samir A. Zaki
- Anatomy Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig 31527, Egypt
| | - Mohammad Algahtani
- Department of Laboratory & Blood Bank, Security Forces Hospital, Mecca P.O. Box 14799, Saudi Arabia
| | - Abdulrahman Theyab
- Department of Laboratory & Blood Bank, Security Forces Hospital, Mecca P.O. Box 14799, Saudi Arabia
- College of Medicine, Al-Faisal University, Riyadh P.O. Box 50927, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah P.O. Box 6231, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence: (M.A.E.); (B.K.)
| |
Collapse
|
14
|
Ozugergin I, Piekny A. Diversity is the spice of life: An overview of how cytokinesis regulation varies with cell type. Front Cell Dev Biol 2022; 10:1007614. [PMID: 36420142 PMCID: PMC9676254 DOI: 10.3389/fcell.2022.1007614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/24/2022] [Indexed: 09/01/2023] Open
Abstract
Cytokinesis is required to physically cleave a cell into two daughters at the end of mitosis. Decades of research have led to a comprehensive understanding of the core cytokinesis machinery and how it is regulated in animal cells, however this knowledge was generated using single cells cultured in vitro, or in early embryos before tissues develop. This raises the question of how cytokinesis is regulated in diverse animal cell types and developmental contexts. Recent studies of distinct cell types in the same organism or in similar cell types from different organisms have revealed striking differences in how cytokinesis is regulated, which includes different threshold requirements for the structural components and the mechanisms that regulate them. In this review, we highlight these differences with an emphasis on pathways that are independent of the mitotic spindle, and operate through signals associated with the cortex, kinetochores, or chromatin.
Collapse
Affiliation(s)
- Imge Ozugergin
- Department of Biology, McGill University, Montreal, QC, Canada
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Alisa Piekny
- Department of Biology, Concordia University, Montreal, QC, Canada
| |
Collapse
|
15
|
Husser MC, Ozugergin I, Resta T, Martin VJJ, Piekny AJ. Cytokinetic diversity in mammalian cells is revealed by the characterization of endogenous anillin, Ect2 and RhoA. Open Biol 2022; 12:220247. [PMID: 36416720 PMCID: PMC9683116 DOI: 10.1098/rsob.220247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cytokinesis is required to physically separate the daughter cells at the end of mitosis. This crucial process requires the assembly and ingression of an actomyosin ring, which must occur with high fidelity to avoid aneuploidy and cell fate changes. Most of our knowledge of mammalian cytokinesis was generated using over-expressed transgenes in HeLa cells. Over-expression can introduce artefacts, while HeLa are cancerous human cells that have lost their epithelial identity, and the mechanisms controlling cytokinesis in these cells could be vastly different from other cell types. Here, we tagged endogenous anillin, Ect2 and RhoA with mNeonGreen and characterized their localization during cytokinesis for the first time in live human cells. Comparing anillin localization in multiple cell types revealed cytokinetic diversity with differences in the duration and symmetry of ring closure, and the timing of cortical recruitment. Our findings show that the breadth of anillin correlates with the rate of ring closure, and support models where cell size or ploidy affects the cortical organization, and intrinsic mechanisms control the symmetry of ring closure. This work highlights the need to study cytokinesis in more diverse cell types, which will be facilitated by the reagents generated for this study.
Collapse
Affiliation(s)
| | - Imge Ozugergin
- Biology Department, Concordia University, Montreal, Quebec, Canada
| | - Tiziana Resta
- Biology Department, Concordia University, Montreal, Quebec, Canada
| | - Vincent J. J. Martin
- Biology Department, Concordia University, Montreal, Quebec, Canada,Center for Applied Synthetic Biology, Concordia University, Montreal, Quebec, Canada
| | - Alisa J. Piekny
- Biology Department, Concordia University, Montreal, Quebec, Canada,Center for Applied Synthetic Biology, Concordia University, Montreal, Quebec, Canada,Center for Microscopy and Cellular Imaging, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Sharma A, Yadav D, Rao P, Sinha S, Goswami D, Rawal RM, Shrivastava N. Identification of potential therapeutic targets associated with diagnosis and prognosis of colorectal cancer patients based on integrated bioinformatics analysis. Comput Biol Med 2022; 146:105688. [DOI: 10.1016/j.compbiomed.2022.105688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 01/04/2023]
|
17
|
Chen J, Li Z, Jia X, Song W, Wu H, Zhu H, Xuan Z, Du Y, Zhu X, Song G, Dong H, Bian S, Wang S, Zhao Y, Xie H, Zheng S, Song P. Targeting anillin inhibits tumorigenesis and tumor growth in hepatocellular carcinoma via impairing cytokinesis fidelity. Oncogene 2022; 41:3118-3130. [PMID: 35477750 DOI: 10.1038/s41388-022-02274-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 11/09/2022]
Abstract
Targeting cytokinesis can suppress tumor growth by blocking cell division and promoting apoptosis. We aimed to characterize key cytokinesis regulator in hepatocellular carcinoma (HCC) progression, providing insights into identifying promising HCC therapeutic targets. The unbiased bioinformatic screening identified Anillin actin binding protein (ANLN) as a critical cytokinesis regulator involved in HCC development. Functional assay demonstrated that knockdown of ANLN inhibited HCC growth by inducing cytokinesis failure and DNA damage, leading to multinucleation and mitotic catastrophe. Mechanistically, ANLN acts as a scaffold to strengthen interaction between RACGAP1 and PLK1. ANLN promotes PLK1-mediated RACGAP1 phosphorylation and RhoA activation to ensure cytokinesis fidelity. To explore the function of ANLN in HCC tumorigenesis, we hydrodynamically transfected c-Myc and NRAS plasmids into Anln+/+, Anln+/-, and Anln-/- mice through tail vein injection. Hepatic Anln ablation significantly impaired c-Myc/NRAS-driven hepatocarcinogenesis. Moreover, enhanced hepatic polyploidization was observed in Anln ablation mice, manifesting as increasing proportion of cellular and nuclear polyploidy. Clinically, ANLN is upregulated in human HCC tissues and high level of ANLN is correlated with poor patients' prognosis. Additionally, the proportion of cellular polyploidy decreases during HCC progression and ANLN level is significantly correlated with cellular polyploidy proportion in human HCC samples. In conclusion, ANLN is identified as a key cytokinesis regulator contributing to HCC initiation and progression. Our findings revealed a novel mechanism of ANLN in the regulation of cytokinesis to promote HCC tumorigenesis and growth, suggesting targeting ANLN to inhibit cytokinesis may be a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Zequn Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Xing Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Hai Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Zefeng Xuan
- Division of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yehui Du
- Division of Thyroid Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingxin Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Guangyuan Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Haijiang Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Suchen Bian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Shuo Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China.,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China. .,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China. .,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China.
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China. .,Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China. .,Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
18
|
Microtubule and Actin Cytoskeletal Dynamics in Male Meiotic Cells of Drosophila melanogaster. Cells 2022; 11:cells11040695. [PMID: 35203341 PMCID: PMC8870657 DOI: 10.3390/cells11040695] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 01/12/2023] Open
Abstract
Drosophila dividing spermatocytes offer a highly suitable cell system in which to investigate the coordinated reorganization of microtubule and actin cytoskeleton systems during cell division of animal cells. Like male germ cells of mammals, Drosophila spermatogonia and spermatocytes undergo cleavage furrow ingression during cytokinesis, but abscission does not take place. Thus, clusters of primary and secondary spermatocytes undergo meiotic divisions in synchrony, resulting in cysts of 32 secondary spermatocytes and then 64 spermatids connected by specialized structures called ring canals. The meiotic spindles in Drosophila males are substantially larger than the spindles of mammalian somatic cells and exhibit prominent central spindles and contractile rings during cytokinesis. These characteristics make male meiotic cells particularly amenable to immunofluorescence and live imaging analysis of the spindle microtubules and the actomyosin apparatus during meiotic divisions. Moreover, because the spindle assembly checkpoint is not robust in spermatocytes, Drosophila male meiosis allows investigating of whether gene products required for chromosome segregation play additional roles during cytokinesis. Here, we will review how the research studies on Drosophila male meiotic cells have contributed to our knowledge of the conserved molecular pathways that regulate spindle microtubules and cytokinesis with important implications for the comprehension of cancer and other diseases.
Collapse
|
19
|
Liu L, Dai X, Yin S, Liu P, Hill EG, Wei W, Gan W. DNA-PK promotes activation of the survival kinase AKT in response to DNA damage through an mTORC2-ECT2 pathway. Sci Signal 2022; 15:eabh2290. [PMID: 34982576 DOI: 10.1126/scisignal.abh2290] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Liu Liu
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA.,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Xiaoming Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shasha Yin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA.,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center and Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth G Hill
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenjian Gan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA.,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
20
|
Li L, Liu S, Peng L, Zhang Y, Zhang Y, Zeng H, Li G, Zhang C. The identification and preliminary study of lncRNA TUG1 and its related genes in hepatocellular carcinoma. Arch Med Sci 2022; 18:1582-1595. [PMID: 36457956 PMCID: PMC9710294 DOI: 10.5114/aoms.2019.89707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/31/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a common malignant tumour of the digestive system, which is a threat to public health. The purpose of this study was to investigate the featured genes and pathways of HCC from a bioinformatics database, and verify their correlation with diagnosis and prognosis of HCC. MATERIAL AND METHODS We downloaded the gene expression profile on HCC from the Gene Expression Omnibus (GEO), and software R was used to identify differentially expressed lncRNA (DEL). The target genes of the lncRNA were further predicted by using a cluster database and molecular interaction database. Subsequently, a combined interaction network of target genes was constructed using the Cytoscape platform with preliminary verification at the level of different databases, cell lines, and tissues. Finally, we explored the effectiveness of TUG1 and its target genes on the diagnosis and prognosis of HCC by univariate Cox analysis and survival analysis. RESULTS A total of four DELs were identified and the most remarkably up-regulated lncRNA was TUG1, which included 12 high-confidence target genes. Moreover, we found that the expression changes of TUG1 and its target genes in different databases, cell lines, and liver cancer tissues were consistent with the prediction. The high expression of TUG1 and its target genes could significantly predict the shorter survival time of HCC patients, among which NCAPG, MCM6, PIGC, PEA15, and RACGAP1 have significant diagnostic value for HCC (AUC > 0.9). CONCLUSIONS This study provides a starting point for the screening of therapeutically relevant targets in HCC. Further experiment should be conducted to verify our findings.
Collapse
Affiliation(s)
- Lei Li
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- School of Basic Medical Science, Central South University, Changsha, China
| | - Shuiping Liu
- School of Basic Medical Science, Central South University, Changsha, China
| | - Li Peng
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yapeng Zhang
- School of Basic Medical Science, Central South University, Changsha, China
| | - Yaqin Zhang
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
| | - Haiyan Zeng
- School of Basic Medical Science, Central South University, Changsha, China
| | - Guancheng Li
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
| | - Chaoyang Zhang
- Cancer Research Institute, Central South University, Changsha, China
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
21
|
Longhini KM, Glotzer M. Aurora A and cortical flows promote polarization and cytokinesis by inducing asymmetric ECT-2 accumulation. eLife 2022; 11:83992. [PMID: 36533896 PMCID: PMC9799973 DOI: 10.7554/elife.83992] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
In the early Caenorhabditis elegans embryo, cell polarization and cytokinesis are interrelated yet distinct processes. Here, we sought to understand a poorly understood aspect of cleavage furrow positioning. Early C. elegans embryos deficient in the cytokinetic regulator centralspindlin form furrows, due to an inhibitory activity that depends on aster positioning relative to the polar cortices. Here, we show polar relaxation is associated with depletion of cortical ECT-2, a RhoGEF, specifically at the posterior cortex. Asymmetric ECT-2 accumulation requires intact centrosomes, Aurora A (AIR-1), and myosin-dependent cortical flows. Within a localization competent ECT-2 fragment, we identified three putative phospho-acceptor sites in the PH domain of ECT-2 that render ECT-2 responsive to inhibition by AIR-1. During both polarization and cytokinesis, our results suggest that centrosomal AIR-1 breaks symmetry via ECT-2 phosphorylation; this local inhibition of ECT-2 is amplified by myosin-driven flows that generate regional ECT-2 asymmetry. Together, these mechanisms cooperate to induce polarized assembly of cortical myosin, contributing to both embryo polarization and cytokinesis.
Collapse
Affiliation(s)
- Katrina M Longhini
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicagoUnited States
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicagoUnited States
| |
Collapse
|
22
|
Wei L, Wang Y, Zhou D, Li X, Wang Z, Yao G, Wang X. Bioinformatics analysis on enrichment analysis of potential hub genes in breast cancer. Transl Cancer Res 2021; 10:2399-2408. [PMID: 35116555 PMCID: PMC8797715 DOI: 10.21037/tcr-21-749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/21/2021] [Indexed: 11/07/2022]
Abstract
Background Despite recent advances in screening, treatment, and survival, breast cancer remains the most invasive cancer in women. The development of novel diagnostic and therapeutic markers for breast cancer may provide more information about its pathogenesis and progression. Methods We obtained GSE86374 micro-expression matrix chip data from the Gene Expression Omnibus (GEO) database consisting of 159 samples (124 normal samples and 35 breast cancer samples). The language was then used to perform data processing and differential expression analysis. For all differentially expressed genes (DEGs), “FDR <0.01 and |logFC| ≥1” were selected as thresholds. Results In this study, 173 up-regulated genes and 143 down-regulated genes were selected for GO and KEGG enrichment analysis. These genes are also significantly enriched in the KEGG pathway, including phenylalanine metabolism, staphylococcus aureus infection, and the PPAR signaling pathway. The survival and prognosis of the selected eight key genes (DLGAP5, PRC1, TOP2A, CENPF, RACGAP1, RRM2, PLK1, and ASPM) were analyzed by the Kaplan-Meier plotter database. Conclusions Eight hub genes and pathways closely related to the onset and progression of breast cancer were identified. We found that the PPAR signaling pathway, especially PPARγ, plays an important role in breast cancer and suggest this pathway be the subject of further research.
Collapse
Affiliation(s)
- Limin Wei
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Yukun Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Dan Zhou
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Xinyang Li
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Ziming Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Ge Yao
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Xinshuai Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
23
|
Schneid S, Wolff F, Buchner K, Bertram N, Baygün S, Barbosa P, Mangal S, Zanin E. The BRCT domains of ECT2 have distinct functions during cytokinesis. Cell Rep 2021; 34:108805. [PMID: 33657383 DOI: 10.1016/j.celrep.2021.108805] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 12/18/2020] [Accepted: 02/08/2021] [Indexed: 12/28/2022] Open
Abstract
During cell division, the guanine nucleotide exchange factor (GEF) ECT2 activates RhoA in a narrow zone at the cell equator in anaphase. ECT2 consists of three BRCT domains (BRCT0, 1, and 2), a catalytic GEF, and a pleckstrin homology (PH) domain. How the conserved BRCT domains spatially and temporally control ECT2 activity remains unclear. We reveal that each BRCT domain makes distinct contributions to the ECT2 function. We find that BRCT0 contributes to, and BRCT1 is essential for, ECT2 activation in anaphase. BRCT2 integrates two functions: GEF inhibition and RACGAP1 binding, which together limit ECT2 activity to a narrow zone at the cell equator. BRCT2-dependent control of active RhoA zone dimension functions in addition to the inhibitory signal of the astral microtubules. Our analysis provides detailed mechanistic insights into how ECT2 activity is regulated and how that regulation ensures, together with other signaling pathways, successful cell division.
Collapse
Affiliation(s)
- Sandra Schneid
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany
| | - Friederike Wolff
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany
| | - Kristina Buchner
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany
| | - Nils Bertram
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany
| | - Seren Baygün
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany
| | - Pedro Barbosa
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany
| | - Sriyash Mangal
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany
| | - Esther Zanin
- Department Biology II, Ludwig-Maximilians University, Planegg-Martinsried, Munich 82152, Germany.
| |
Collapse
|
24
|
Ren K, Zhou D, Wang M, Li E, Hou C, Su Y, Zou Q, Zhou P, Liu X. RACGAP1 modulates ECT2-Dependent mitochondrial quality control to drive breast cancer metastasis. Exp Cell Res 2021; 400:112493. [PMID: 33485843 DOI: 10.1016/j.yexcr.2021.112493] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023]
Abstract
Most cancer deaths are due to the colonization of tumor cells in distant organs. More evidence indicates that overexpression of RACGAP1 plays a critical role in cancer metastasis. However, the underlying mechanism still remains poorly understood. Here we found that RACGAP1 promoted breast cancer metastasis through regulating mitochondrial quality control. Overexpression of RACGAP1 in breast cancer cells led to the fragmentation of mitochondria, increased mitophagy intensity, mitochondrial turnover, and aerobic glycolysis ATP production. We showed that RACGAP1 promoted mitochondrial fission through recruiting ECT2 during anaphase and subsequently had activated ERK-DRP1 pathway. We further demonstrated the phosphorylation of RACGAP1 is essential for its ability of binding with ECT2 and its downstream effects. RACGAP1 overexpression also increased the expression of PGC-1a, a key mitochondrial biogenesis regulator, presumably by the increased mitophagy intensity induced by RACGAP1. PGC-1a increased the enrichment of DNMT1 in mitochondria, mitochondrial DNMT1 augmented mitochondrial DNA methylation and upregulated mitochondrial genome transcription. Our data indicated that RACGAP1 simultaneously facilitated mitophagy and mitochondrial biogenesis through regulating DRP1 phosphorylation and PGC-1a expression, eventually improved mitochondrial quality control in breast cancer cells. Our study provided a new angle in understanding the RACGAP1-overexpression related malignancy in breast cancer patients.
Collapse
Affiliation(s)
- Kehan Ren
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Danmei Zhou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Meili Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China; Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Ermin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenjian Hou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ying Su
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qiang Zou
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Ping Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiuping Liu
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
| |
Collapse
|
25
|
Huang C, Hu CG, Ning ZK, Huang J, Zhu ZM. Identification of key genes controlling cancer stem cell characteristics in gastric cancer. World J Gastrointest Surg 2020; 12:442-459. [PMID: 33304447 PMCID: PMC7701879 DOI: 10.4240/wjgs.v12.i11.442] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/13/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Self-renewal of gastric cancer stem cells (GCSCs) is considered to be the underlying cause of the metastasis, drug resistance, and recurrence of gastric cancer (GC).
AIM To characterize the expression of stem cell-related genes in GC.
METHODS RNA sequencing results and clinical data for gastric adenoma and adenocarcinoma samples were obtained from The Cancer Genome Atlas database, and the results of the GC mRNA expression-based stemness index (mRNAsi) were analyzed. Weighted gene coexpression network analysis was then used to find modules of interest and their key genes. Survival analysis of key genes was performed using the online tool Kaplan-Meier Plotter, and the online database Oncomine was used to assess the expression of key genes in GC.
RESULTS mRNAsi was significantly upregulated in GC tissues compared to normal gastric tissues (P < 0.0001). A total of 16 modules were obtained from the gene coexpression network; the brown module was most positively correlated with mRNAsi. Sixteen key genes (BUB1, BUB1B, NCAPH, KIF14, RACGAP1, RAD54L, TPX2, KIF15, KIF18B, CENPF, TTK, KIF4A, SGOL2, PLK4, XRCC2, and C1orf112) were identified in the brown module. The functional and pathway enrichment analyses showed that the key genes were significantly enriched in the spindle cellular component, the sister chromatid segregation biological process, the motor activity molecular function, and the cell cycle and homologous recombination pathways. Survival analysis and Oncomine analysis revealed that the prognosis of patients with GC and the expression of three genes (RAD54L, TPX2, and XRCC2) were consistently related.
CONCLUSION Sixteen key genes are primarily associated with stem cell self-renewal and cell proliferation characteristics. RAD54L, TPX2, and XRCC2 are the most likely therapeutic targets for inhibiting the stemness characteristics of GC cells.
Collapse
Affiliation(s)
- Chao Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Ce-Gui Hu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zhi-Kun Ning
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Jun Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zheng-Ming Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
26
|
Sechi S, Frappaolo A, Karimpour-Ghahnavieh A, Fraschini R, Giansanti MG. A novel coordinated function of Myosin II with GOLPH3 controls centralspindlin localization during cytokinesis in Drosophila. J Cell Sci 2020; 133:jcs252965. [PMID: 33037125 DOI: 10.1242/jcs.252965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
In animal cell cytokinesis, interaction of non-muscle myosin II (NMII) with F-actin provides the dominant force for pinching the mother cell into two daughters. Here we demonstrate that celibe (cbe) is a missense allele of zipper, which encodes the Drosophila Myosin heavy chain. Mutation of cbe impairs binding of Zipper protein to the regulatory light chain Spaghetti squash (Sqh). In dividing spermatocytes from cbe males, Sqh fails to concentrate at the equatorial cortex, resulting in thin actomyosin rings that are unable to constrict. We show that cbe mutation impairs localization of the phosphatidylinositol 4-phosphate [PI(4)P]-binding protein Golgi phosphoprotein 3 (GOLPH3, also known as Sauron) and maintenance of centralspindlin at the cell equator of telophase cells. Our results further demonstrate that GOLPH3 protein associates with Sqh and directly binds the centralspindlin subunit Pavarotti. We propose that during cytokinesis, the reciprocal dependence between Myosin and PI(4)P-GOLPH3 regulates centralspindlin stabilization at the invaginating plasma membrane and contractile ring assembly.
Collapse
Affiliation(s)
- Stefano Sechi
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Anna Frappaolo
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Angela Karimpour-Ghahnavieh
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Roberta Fraschini
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano Bicocca, 20126, Milano, Italy
| | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| |
Collapse
|
27
|
Zhao W, Wang M, Wang C, Liu Y, Liu H, Luo S. RACGAP1 is transcriptionally regulated by E2F3, and its depletion leads to mitotic catastrophe in esophageal squamous cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:950. [PMID: 32953750 PMCID: PMC7475413 DOI: 10.21037/atm-20-2901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background RACGAP1 has significant involvement in tumorigenesis of cancers, including liver cancer, stomach cancer, and colon cancer. However, the role and the exact mechanism of RACGAP1 in esophageal squamous cell carcinoma (ESCC) has not been explored. Methods QPCR and Western blots analysis was performed to analyze the expression of RACGAP1 in ESCC. MTT assays and colony formation assays were performed to explore the functional role of RACGAP1 in ESCC. Cell cycle analysis and immunofluorescence assays were used to investigate the function of RACGAP1 involvement in mitotic catastrophe. At last, we conducted the public datasets mining to explore the expression status and prognosis value of RACGAP1 as well as the correlation between RACGAP1 and E2F3 in various cancers. Results The high abnormal expression of RACGAP1 is observed in ESCC and associated with worse clinical outcomes of patients with ESCC. RACGAP1, a novel cell cycle associated gene regulated by E2F3, acts as an oncogenic driver in ESCC cell lines. Notably, for the first time, RACGAP1 depletion induced severe mitotic catastrophe, followed by massive cell death. Conclusions Our findings showed the essential role of RACGAP1 in ESCC cancer cell survival and the therapeutic potential of RACGAP1 as a molecular target for ESCC.
Collapse
Affiliation(s)
- Weifeng Zhao
- Department of Medical Oncology, the Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,Department of Oncology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Mengyao Wang
- Radiation Oncology Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Chaojie Wang
- Department of Oncology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yingjun Liu
- Department of General Surgery, the Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Huimin Liu
- Department of Oncology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Suxia Luo
- Department of Medical Oncology, the Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
28
|
Zhou W, Zhao S, Xu S, Sun Z, Liang Y, Ding X. RacGAP1 ameliorates acute kidney injury by promoting proliferation and suppressing apoptosis of renal tubular cells. Biochem Biophys Res Commun 2020; 527:624-630. [PMID: 32423815 DOI: 10.1016/j.bbrc.2020.04.140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/27/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) remains correlated with high mortality. Novel therapeutic strategies are urgently needed for AKI patients. Rac GTPase-activating protein 1 (RacGAP1) regulates the activity of RhoGTPase and acts as a predictive biomarker in several types of malignant tumor but the role of RacGAP1 in AKI has not been revealed. METHODS Animal models of AKI induced by renal ischemia-reperfusion (I/R) and cisplatin treatment were generated in C57BL/6 mice. Hypoxia/reoxygenation (H/R) and cisplatin treatment were practiced in human renal tubular epithelial (HK-2) and renal tubular duct epithelial cells of rat (NRK-52E) cells. The role of RacGAP1 in cell proliferation and apoptosis was estimated using western bolting, immunocytochemistry and flow cytometry. Verteporfin was used to activate the Hippo pathway to show whether the protective effects of RacGAP1 on cell growth and survival in renal tubular cells were dependent on the activation of YAP. RESULTS The expression of RacGAP1 was significantly increased in mice kidneys after I/R or cisplatin treatment, combined with increased expression of RacGAP1 in H/R or cisplatin challenged cells. Overexpression of RacGAP1 protected HK2 and NRK-52E cells by promoting proliferation and decreasing apoptosis. We also disclosed that RacGAP1 exerted its function through activation of YAP. CONCLUSION The present study provides evidence that RacGAP1 is involved in AKI. It promotes proliferation and limits apoptosis of tubular epithelial cells via stimulating activation and nuclear translocation of YAP. Consequently, RacGAP1 may be a novel therapeutic target for AKI.
Collapse
Affiliation(s)
- Weiran Zhou
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuan Zhao
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Medical Center of Kidney Disease, Shanghai, China; Shanghai Institute of Kidney and Dialysis, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China; Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Sujuan Xu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaoxing Sun
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiran Liang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Medical Center of Kidney Disease, Shanghai, China; Shanghai Institute of Kidney and Dialysis, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China; Hemodialysis Quality Control Center of Shanghai, Shanghai, China.
| |
Collapse
|
29
|
Beaudet D, Pham N, Skaik N, Piekny A. Importin binding mediates the intramolecular regulation of anillin during cytokinesis. Mol Biol Cell 2020; 31:1124-1139. [PMID: 32238082 PMCID: PMC7353161 DOI: 10.1091/mbc.e20-01-0006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cytokinesis occurs by the ingression of an actomyosin ring that cleaves a cell into two daughters. This process is tightly controlled to avoid aneuploidy, and we previously showed that active Ran coordinates ring positioning with chromatin. Active Ran is high around chromatin, and forms an inverse gradient to cargo-bound importins. We found that the ring component anillin contains a nuclear localization signal (NLS) that binds to importin and is required for its function during cytokinesis. Here we reveal the mechanism whereby importin binding favors a conformation required for anillin's recruitment to the equatorial cortex. Active RhoA binds to the RhoA-binding domain causing an increase in accessibility of the nearby C2 domain containing the NLS. Importin binding subsequently stabilizes a conformation that favors interactions for cortical recruitment. In addition to revealing a novel mechanism for the importin-mediated regulation of a cortical protein, we also show how importin binding positively regulates protein function.
Collapse
Affiliation(s)
- Daniel Beaudet
- Department of Bioengineering, McGill University, Montreal, QC, Canada, H3A 0G4
| | - Nhat Pham
- Department of Biology, Concordia University, Montreal, QC, Canada, H4B 1R6
| | - Noha Skaik
- Department of Biology, Concordia University, Montreal, QC, Canada, H4B 1R6
| | - Alisa Piekny
- Department of Biology, Concordia University, Montreal, QC, Canada, H4B 1R6
| |
Collapse
|
30
|
Papini D, Fant X, Ogawa H, Desban N, Samejima K, Feizbakhsh O, Askin B, Ly T, Earnshaw WC, Ruchaud S. Cell cycle-independent furrowing triggered by phosphomimetic mutations of the INCENP STD motif requires Plk1. J Cell Sci 2019; 132:jcs234401. [PMID: 31601613 PMCID: PMC7115952 DOI: 10.1242/jcs.234401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 11/20/2022] Open
Abstract
Timely and precise control of Aurora B kinase, the chromosomal passenger complex (CPC) catalytic subunit, is essential for accurate chromosome segregation and cytokinesis. Post-translational modifications of CPC subunits are directly involved in controlling Aurora B activity. Here, we identified a highly conserved acidic STD-rich motif of INCENP that is phosphorylated during mitosis in vivo and by Plk1 in vitro and is involved in controlling Aurora B activity. By using an INCENP conditional-knockout cell line, we show that impairing the phosphorylation status of this region disrupts chromosome congression and induces cytokinesis failure. In contrast, mimicking constitutive phosphorylation not only rescues cytokinesis but also induces ectopic furrows and contractile ring formation in a Plk1- and ROCK1-dependent manner independent of cell cycle and microtubule status. Our experiments identify the phospho-regulation of the INCENP STD motif as a novel mechanism that is key for chromosome alignment and cytokinesis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Diana Papini
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
| | - Xavier Fant
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
- Sorbonne Université/CNRS UMR8227, Station Biologique, Place Georges Teissier, CS90074, 29688 ROSCOFF cedex, France
| | - Hiromi Ogawa
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
| | - Nathalie Desban
- Sorbonne Université/CNRS UMR8227, Station Biologique, Place Georges Teissier, CS90074, 29688 ROSCOFF cedex, France
| | - Kumiko Samejima
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
| | - Omid Feizbakhsh
- Sorbonne Université/CNRS UMR8227, Station Biologique, Place Georges Teissier, CS90074, 29688 ROSCOFF cedex, France
| | - Bilge Askin
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
| | - Tony Ly
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
| | - William C. Earnshaw
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
| | - Sandrine Ruchaud
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, Scotland, UK
- Sorbonne Université/CNRS UMR8227, Station Biologique, Place Georges Teissier, CS90074, 29688 ROSCOFF cedex, France
| |
Collapse
|
31
|
Abstract
The active form of the small GTPase RhoA is necessary and sufficient for formation of a cytokinetic furrow in animal cells. Despite the conceptual simplicity of the process, the molecular mechanisms that control it are intricate and involve redundancy at multiple levels. Here, we discuss our current knowledge of the mechanisms underlying spatiotemporal regulation of RhoA during cytokinesis by upstream activators. The direct upstream activator, the RhoGEF Ect2, requires activation due to autoinhibition. Ect2 is primarily activated by the centralspindlin complex, which contains numerous domains that regulate its subcellular localization, oligomeric state, and Ect2 activation. We review the functions of these domains and how centralspindlin is regulated to ensure correctly timed, equatorial RhoA activation. Highlighting recent evidence, we propose that although centralspindlin does not always prominently accumulate on the plasma membrane, it is the site where it promotes RhoA activation during cytokinesis.
Collapse
|
32
|
Yin C, Toiyama Y, Okugawa Y, Shigemori T, Yamamoto A, Ide S, Kitajima T, Fujikawa H, Yasuda H, Okita Y, Hiro J, Yoshiyama S, Ohi M, Araki T, Yao L, Kusunoki M. Rac GTPase-Activating Protein 1 (RACGAP1) as an Oncogenic Enhancer in Esophageal Carcinoma. Oncology 2019; 97:155-163. [PMID: 31216559 DOI: 10.1159/000500592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/24/2019] [Indexed: 11/19/2022]
Abstract
PURPOSE Rac GTPase-activating protein 1 (RACGAP1) is associated with cell proliferation, and there is much evidence of its oncogenic role. This study investigated the clinical importance and functional role of RACGAP1 in esophageal carcinoma (EC). METHODS A total of 81 EC patients were enrolled in the study. We assessed the immunohistochemical score of EC tissues and adjacent normal esophageal mucosae, and then performed multiple cell function tests by means of in vitro experiments to elucidate the functional role of RACGAP1 using RNA interference technology in EC cell lines. RESULTS RACGAP1 was significantly overexpressed in EC tissues compared with the adjacent normal esophageal mucosae (p < 0.0001). Moreover, RACGAP1 overexpression was significantly correlated with poor overall survival (p = 0.032) and disease-free survival (p = 0.012) in EC patients. High RACGAP1 expression was also significantly correlated with the presence of lymphatic invasion (p = 0.012), vessel invasion (p = 0.003), and advanced TNM (tumor-node-metastasis) stage (p = 0.046) in EC patients. In vitro analysis demonstrated that RACGAP1 was involved in the proliferation, tumorigenicity, invasion, migration, and anoikis resistance in EC cells. CONCLUSIONS RACGAP1 plays a pivotal role in EC development, suggesting that it could be used as an indicator of prognosis in EC patients.
Collapse
Affiliation(s)
- Chengzeng Yin
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan.,Department of Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yuji Toiyama
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan,
| | - Yoshinaga Okugawa
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Tsunehiko Shigemori
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Akira Yamamoto
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Shozo Ide
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Takahito Kitajima
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiroyuki Fujikawa
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiromi Yasuda
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Yoshiki Okita
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Junichiro Hiro
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Shigeyuki Yoshiyama
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Masaki Ohi
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Toshimitsu Araki
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Li Yao
- Department of Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Masato Kusunoki
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
33
|
Adriaans IE, Basant A, Ponsioen B, Glotzer M, Lens SM. PLK1 plays dual roles in centralspindlin regulation during cytokinesis. J Cell Biol 2019; 218:1250-1264. [PMID: 30728176 PMCID: PMC6446842 DOI: 10.1083/jcb.201805036] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 12/26/2018] [Accepted: 01/23/2019] [Indexed: 11/26/2022] Open
Abstract
Cytokinesis begins upon anaphase onset. An early step involves local activation of the small GTPase RhoA, which triggers assembly of an actomyosin-based contractile ring at the equatorial cortex. Here, we delineated the contributions of PLK1 and Aurora B to RhoA activation and cytokinesis initiation in human cells. Knock-down of PRC1, which disrupts the spindle midzone, revealed the existence of two pathways that can initiate cleavage furrow ingression. One pathway depends on a well-organized spindle midzone and PLK1, while the other depends on Aurora B activity and centralspindlin at the equatorial cortex and can operate independently of PLK1. We further show that PLK1 inhibition sequesters centralspindlin onto the spindle midzone, making it unavailable for Aurora B at the equatorial cortex. We propose that PLK1 activity promotes the release of centralspindlin from the spindle midzone through inhibition of PRC1, allowing centralspindlin to function as a regulator of spindle midzone formation and as an activator of RhoA at the equatorial cortex.
Collapse
Affiliation(s)
- Ingrid E. Adriaans
- Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Angika Basant
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL
| | - Bas Ponsioen
- Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL
| | - Susanne M.A. Lens
- Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
34
|
Yang XM, Cao XY, He P, Li J, Feng MX, Zhang YL, Zhang XL, Wang YH, Yang Q, Zhu L, Nie HZ, Jiang SH, Tian GA, Zhang XX, Liu Q, Ji J, Zhu X, Xia Q, Zhang ZG. Overexpression of Rac GTPase Activating Protein 1 Contributes to Proliferation of Cancer Cells by Reducing Hippo Signaling to Promote Cytokinesis. Gastroenterology 2018; 155:1233-1249.e22. [PMID: 30009820 DOI: 10.1053/j.gastro.2018.07.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Agents designed to block or alter cytokinesis can kill or stop proliferation of cancer cells. We aimed to identify cytokinesis-related proteins that are overexpressed in hepatocellular carcinoma (HCC) cells and might be targeted to slow liver tumor growth. METHODS Using the Oncomine database, we compared the gene expression patterns in 16 cancer microarray datasets and assessed gene enrichment sets using gene ontology. We performed immunohistochemical analysis of an HCC tissue microarray and identified changes in protein levels that are associated with patient survival times. Candidate genes were overexpressed or knocked down with small hairpin RNAs in SMMC7721, MHCC97H, or HCCLM3 cell lines; we analyzed their proliferation, viability, and clone-formation ability and their growth as subcutaneous or orthotopic xenograft tumors in mice. We performed microarray analyses to identify alterations in signaling pathways and immunoblot and immunofluorescence assays to detect and localize proteins in tissues. Yeast 2-hybrid screens and mass spectrometry combined with co-immunoprecipitation experiments were used to identify binding proteins. Protein interactions were validated with co-immunoprecipitation and proximity ligation assays. Chromatin immunoprecipitation, promoter luciferase activity, and quantitative real-time polymerase chain reaction analyses were used to identify factors that regulate transcription of specific genes. RESULTS The genes that were most frequently overexpressed in different types of cancer cells were involved in cell division processes. We identified 3 cytokinesis-regulatory proteins among the 10 genes most frequently overexpressed by all cancer cell types. Rac GTPase activating protein 1 (RACGAP1) was the cytokinesis-regulatory protein that was most highly overexpressed in multiple cancers. Increased expression of RACGAP1 in tumor tissues was associated with shorter survival times of patients with cancer. Knockdown of RACGAP1 in HCC cells induced cytokinesis failure and cell apoptosis. In microarray analyses, we found knockdown of RACGAP1 in SMMC7721 cells to reduce expression of genes regulated by yes-associated protein (YAP) and WW domain containing transcription regulator 1 (WWTR1 or TAZ). RACGAP1 reduced activation of the Hippo pathway in HCC cells by increasing activity of RhoA and polymerization of filamentous actin. Knockdown of YAP reduced phosphorylation of RACGAP1 and redistribution at the anaphase central spindle. We found transcription of the translocated promoter region, nuclear basket protein (TPR) to be regulated by YAP and coordinately expressed with RACGAP1 to promote proliferation of HCC cells. TPR redistributed upon nuclear envelope breakdown and formed complexes with RACGAP1 during mitosis. Knockdown of TPR in HCC cells reduced phosphorylation of RACGAP1 by aurora kinase B and impaired their redistribution at the central spindle during cytokinesis. STAT3 activated transcription of RACGAP in HCC cells. CONCLUSIONS In an analysis of gene expression patterns of multiple tumor types, we found RACGAP1 to be frequently overexpressed, which is associated with shorter survival times of patients. RACGAP1 promotes proliferation of HCC cells by reducing activation of the Hippo and YAP pathways and promoting cytokinesis in coordination with TPR.
Collapse
Affiliation(s)
- Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Yan Cao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping He
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Xuan Feng
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Zhen Nie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-Ang Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Liu
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University Jan Waldenströms gata 35 Skåne University Hospital, Malmö, Sweden
| | - Xuefeng Zhu
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Qiang Xia
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
35
|
Yin X, Sun J, Zhang H, Wang S. Comprehensive analysis of multi Ewing sarcoma microarray datasets identifies several prognosis biomarkers. Mol Med Rep 2018; 18:4229-4238. [PMID: 30221671 PMCID: PMC6172382 DOI: 10.3892/mmr.2018.9432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 07/25/2018] [Indexed: 11/27/2022] Open
Abstract
Ewing sarcoma (ES) is a common primary malignancy in children and adolescents. Progression of treatment methods hasn't contributed a lot to the imrovement of prognosis. To identify potential prognostic biomarkers, a meta-analysis pipeline of multi-gene expression datasets for ES from the Gene Expression Omnibus (GEO) was performed. Three datasets were screened and differential expression genes (DEGs) in ES samples compared with normal tissues were identified through limma package and subjected to network analysis. As a result, 1,470 DEGs were obtained which were mainly involved in biological processes associated with immune response and transcription regulation. Network analysis obtained 22 core genes with high network degree and fold change. Kaplan-Meier analysis based on ES datasets from The Cancer Genome Atlas identified five genes, including glycogen phosphorylase, muscle-associated, myocyte-specific enhancer factor 2C, tripartite motif containing 63, budding uninhibited by benzimidazoses1 and Ras GTPase-activating protein 1, whose altered expression profiles are significantly associated with survival. Changes of their expression values were further confirmed through RT-qPCR in ES cell and normal cell lines. Those genes may be considered as potential prognostic biomarkers of ES and should be helpful for its early diagnosis and treatment.
Collapse
Affiliation(s)
- Xuqing Yin
- Department of Traumatic Orthopaedics, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| | - Jiubo Sun
- Department of Radiation Oncology, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| | - Haiyang Zhang
- Department of Microinvasive Othopaedics, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| | - Shuai Wang
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
36
|
HIPK2 and extrachromosomal histone H2B are separately recruited by Aurora-B for cytokinesis. Oncogene 2018; 37:3562-3574. [PMID: 29563611 PMCID: PMC6021368 DOI: 10.1038/s41388-018-0191-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/24/2018] [Accepted: 02/05/2018] [Indexed: 11/09/2022]
Abstract
Cytokinesis, the final phase of cell division, is necessary to form two distinct daughter cells with correct distribution of genomic and cytoplasmic materials. Its failure provokes genetically unstable states, such as tetraploidization and polyploidization, which can contribute to tumorigenesis. Aurora-B kinase controls multiple cytokinetic events, from chromosome condensation to abscission when the midbody is severed. We have previously shown that HIPK2, a kinase involved in DNA damage response and development, localizes at the midbody and contributes to abscission by phosphorylating extrachromosomal histone H2B at Ser14. Of relevance, HIPK2-defective cells do not phosphorylate H2B and do not successfully complete cytokinesis leading to accumulation of binucleated cells, chromosomal instability, and increased tumorigenicity. However, how HIPK2 and H2B are recruited to the midbody during cytokinesis is still unknown. Here, we show that regardless of their direct (H2B) and indirect (HIPK2) binding of chromosomal DNA, both H2B and HIPK2 localize at the midbody independently of nucleic acids. Instead, by using mitotic kinase-specific inhibitors in a spatio-temporal regulated manner, we found that Aurora-B kinase activity is required to recruit both HIPK2 and H2B to the midbody. Molecular characterization showed that Aurora-B directly binds and phosphorylates H2B at Ser32 while indirectly recruits HIPK2 through the central spindle components MgcRacGAP and PRC1. Thus, among different cytokinetic functions, Aurora-B separately recruits HIPK2 and H2B to the midbody and these activities contribute to faithful cytokinesis.
Collapse
|
37
|
Lee SR, Jo YJ, Namgoong S, Kim NH. Anillin controls cleavage furrow formation in the course of asymmetric division during mouse oocyte maturation. Mol Reprod Dev 2018; 83:792-801. [PMID: 27508507 DOI: 10.1002/mrd.22688] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 08/08/2016] [Indexed: 12/31/2022]
Abstract
Anillin is a scaffold protein that recruits several proteins involved in cleavage furrow formation during cytokinesis. The role of anilllin in symmetric cell divisions in somatic cells has been intensively studied, yet its involvement in cleavage furrow formation is still elusive. In this study, we investigated the role of anillin in mammalian oocyte maturation and cytokinesis. We found that anillin is localized around the nucleus during the oocyte germinal-vesicle stage, and spreads to the cytoplasm after germinal vesicle breakdown. Thereafter, anillin concentrates at the site of the cleavage furrow from anaphase I to metaphase II. Disruption of anillin activity by microinjecting oocytes with specific siRNAs resulted in a failure of polar body extrusion and asymmetric division, and caused abnormal chromosome segregation during anaphase I. Furthermore, pharmacological inhibition of myosin light chain using Y-27632 or ML-7 resulted in decreased anillin expression. Collectively, our data suggest that anillin is an essential intracellular component that maintains the integrity of asymmetric division in mouse oocytes. Mol. Reprod. Dev. 83: 792-801, 2016 © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- So-Rim Lee
- Department of Animal Sciences, Chungbuk National University, Cheong-ju, South Korea
| | - Yu-Jin Jo
- Department of Animal Sciences, Chungbuk National University, Cheong-ju, South Korea
| | - Suk Namgoong
- Department of Animal Sciences, Chungbuk National University, Cheong-ju, South Korea.
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Cheong-ju, South Korea.
| |
Collapse
|
38
|
Li Y, Cai X, Chen B, Gu H, Liu C. Overexpression of epithelial cell transforming 2 protein in colorectal carcinoma predicts a poor prognosis. Exp Ther Med 2017; 14:4862-4868. [PMID: 29109759 PMCID: PMC5663027 DOI: 10.3892/etm.2017.5132] [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] [Received: 01/24/2016] [Accepted: 02/17/2017] [Indexed: 02/07/2023] Open
Abstract
Epithelial cell transforming 2 (Ect2) protein is a member of the human diffuse B-cell lymphoma family of guanine nucleotide exchange factors, which activate the Ras homolog gene family of small GTPases; however, the clinical implications of Ect2 in colorectal carcinoma (CRC) are unclear. The present study aimed to determine the relationship between Ect2 expression and prognosis in patients with CRC. Western blot analysis and immunohistochemistry assays were used to determine the expression of Ect2 in CRC and paired non-cancerous tissues from 66 patients. The correlation between Ect2 expression and clinicopathological parameters was assessed using χ2 tests. Patient survival was determined using the Kaplan-Meier method and log-rank test. Cox regression was used for multivariate analysis of prognostic factors. Results demonstrated that Ect2 protein was highly expressed in human CRC samples [29/45 (64.45%)] and significantly correlated with a poor prognosis (P<0.05). Compared with normal tissues, CRC tissues demonstrated higher expression levels of Ect2 mRNA [44/66 (66.67%)]. In addition, highly-expressed Ect2 was significantly associated with recurrence (P=0.023) and invasion (P=0.008) of CRC. High Ect2 expression levels in patients were associated with poorer overall survival (OS) and disease-free survival (DFS) compared with lower expression levels of Ect2. Based on multivariate analysis, Ect2 overexpression was significantly correlated with OS and DFS (P=0.015 and 0.020, respectively). In conclusion, Ect2 overexpression is an independent and important prognostic factor for OS and DFS in patients with CRC.
Collapse
Affiliation(s)
- Yiming Li
- Division of Breast Surgery, Department of Surgical Oncology, General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China.,Department of General Surgery, 202 Hospital of People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Xiangjun Cai
- Department of General Surgery, 202 Hospital of People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Bo Chen
- Division of Breast Surgery, Department of Surgical Oncology, General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hanbo Gu
- Department of General Surgery, 202 Hospital of People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Caigang Liu
- Division of Breast Surgery, Department of Surgical Oncology, General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
39
|
Abstract
Cytokinesis in metazoan cells is mediated by an actomyosin-based contractile ring that assembles in response to activation of the small GTPase RhoA. The guanine nucleotide exchange factor that activates RhoA during cytokinesis, ECT-2, is highly regulated. In most metazoan cells, with the notable exception of the early
Caenorhabditis elegans embryo, RhoA activation and furrow ingression require the centralspindlin complex. This exception is due to the existence of a parallel pathway for RhoA activation in
C. elegans. Centralspindlin contains CYK-4 which contains a predicted Rho family GTPase-activating protein (GAP) domain. The function of this domain has been the subject of considerable debate. Some publications suggest that the GAP domain promotes RhoA activation (for example, Zhang and Glotzer, 2015; Loria, Longhini and Glotzer, 2012), whereas others suggest that it functions to inactivate the GTPase Rac1 (for example, Zhuravlev
et al., 2017). Here, we review the mechanisms underlying RhoA activation during cytokinesis, primarily focusing on data in
C. elegans. We highlight the importance of considering the parallel pathway for RhoA activation and detailed analyses of
cyk-4 mutant phenotypes when evaluating the role of the GAP domain of CYK-4.
Collapse
Affiliation(s)
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
40
|
Beaudet D, Akhshi T, Phillipp J, Law C, Piekny A. Active Ran regulates anillin function during cytokinesis. Mol Biol Cell 2017; 28:3517-3531. [PMID: 28931593 PMCID: PMC5683762 DOI: 10.1091/mbc.e17-04-0253] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 11/11/2022] Open
Abstract
We describe a novel mechanism by which active Ran regulates anillin during cytokinesis. Anillin is highly conserved and coordinates RhoA, actomyosin, microtubules, and the membrane for cytokinesis in mammalian cells. This study implicates Ran-GTP in influencing cortical contractility during anaphase by regulating anillin function. Cytokinesis cleaves a cell into two daughters at the end of mitosis, and must be spatially coordinated with chromosome segregation to prevent aneuploidy. The dogma is that the mitotic spindle governs the assembly and constriction of an actomyosin ring. Here, we reveal a function for active Ran in spatially restricting the ring. Our model is that during anaphase, “free” importins, whose gradient inversely correlates with active Ran and chromatin position, function as a molecular ruler for the recruitment and localization of anillin, a contractile protein and a crucial regulator of cytokinesis. We found that decreasing Ran-GTP levels or tethering active Ran to the equatorial membrane affects anillin’s localization and causes cytokinesis phenotypes. Anillin contains a conserved nuclear localization signal (NLS) at its C-terminus that binds to importin-β and is required for cortical polarity and cytokinesis. Mutating the NLS decreases anillin’s cortical affinity, causing it to be more dominantly regulated by microtubules. Anillin contains a RhoA-GTP binding domain, which autoinhibits the NLS and the neighboring microtubule-binding domain, and RhoA-GTP binding may relieve this inhibition during mitosis. Retention of the C-terminal NLS in anillin homologues suggests that this is a conserved mechanism for controlling anillin function.
Collapse
Affiliation(s)
- Daniel Beaudet
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Tara Akhshi
- Program in Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Julia Phillipp
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Christopher Law
- Centre for Microscopy and Cellular Imaging, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Alisa Piekny
- Program in Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
41
|
de Cárcer G, Wachowicz P, Martínez-Martínez S, Oller J, Méndez-Barbero N, Escobar B, González-Loyola A, Takaki T, El Bakkali A, Cámara JA, Jiménez-Borreguero LJ, Bustelo XR, Cañamero M, Mulero F, de Los Ángeles Sevilla M, Montero MJ, Redondo JM, Malumbres M. Plk1 regulates contraction of postmitotic smooth muscle cells and is required for vascular homeostasis. Nat Med 2017; 23:964-974. [PMID: 28692064 DOI: 10.1038/nm.4364] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 06/13/2017] [Indexed: 12/19/2022]
Abstract
Polo-like kinase 1 (PLK1), an essential regulator of cell division, is currently undergoing clinical evaluation as a target for cancer therapy. We report an unexpected function of Plk1 in sustaining cardiovascular homeostasis. Plk1 haploinsufficiency in mice did not induce obvious cell proliferation defects but did result in arterial structural alterations, which frequently led to aortic rupture and death. Specific ablation of Plk1 in vascular smooth muscle cells (VSMCs) led to reduced arterial elasticity, hypotension, and an impaired arterial response to angiotensin II in vivo. Mechanistically, we found that Plk1 regulated angiotensin II-dependent activation of RhoA and actomyosin dynamics in VSMCs in a mitosis-independent manner. This regulation depended on Plk1 kinase activity, and the administration of small-molecule Plk1 inhibitors to angiotensin II-treated mice led to reduced arterial fitness and an elevated risk of aneurysm and aortic rupture. We thus conclude that a partial reduction of Plk1 activity that does not block cell division can nevertheless impair aortic homeostasis. Our findings have potentially important implications for current approaches aimed at PLK1 inhibition for cancer therapy.
Collapse
Affiliation(s)
- Guillermo de Cárcer
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Paulina Wachowicz
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sara Martínez-Martínez
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
| | - Jorge Oller
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
| | - Nerea Méndez-Barbero
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
| | - Beatriz Escobar
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Tohru Takaki
- Clare Hall Laboratories, London Research Institute, London, UK
| | - Aicha El Bakkali
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Juan A Cámara
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Luis J Jiménez-Borreguero
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
- Advanced Imaging Unit, Spanish National Cardiovascular Centre (CNIC), and Cardiac Imaging Department, Hospital de la Princesa, Madrid, Spain
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer de Salamanca, University of Salamanca-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Marta Cañamero
- Comparative Pathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - María de Los Ángeles Sevilla
- Department of Physiology and Pharmacology and Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - María Jose Montero
- Department of Physiology and Pharmacology and Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
| | - Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
42
|
Zhuravlev Y, Hirsch SM, Jordan SN, Dumont J, Shirasu-Hiza M, Canman JC. CYK-4 regulates Rac, but not Rho, during cytokinesis. Mol Biol Cell 2017; 28:1258-1270. [PMID: 28298491 PMCID: PMC5415020 DOI: 10.1091/mbc.e17-01-0020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/23/2017] [Accepted: 03/02/2017] [Indexed: 12/18/2022] Open
Abstract
The roles of the Rho-family GAP CYK-4 and small GTPase Rac during cytokinesis are examined in Caenorhabditis elegans embryos. CYK-4 opposes Rac (and potentially Cdc42) activity during cytokinesis. There is no evidence that CYK-4 is upstream of Rho activity or that Rac disruption is a general suppressor of cytokinesis failure. Cytokinesis is driven by constriction of an actomyosin contractile ring that is controlled by Rho-family small GTPases. Rho, activated by the guanine-nucleotide exchange factor ECT-2, is upstream of both myosin-II activation and diaphanous formin-mediated filamentous actin (f-actin) assembly, which drive ring constriction. The role for Rac and its regulators is more controversial, but, based on the finding that Rac inactivation can rescue cytokinesis failure when the GTPase-activating protein (GAP) CYK-4 is disrupted, Rac activity was proposed to be inhibitory to contractile ring constriction and thus specifically inactivated by CYK-4 at the division plane. An alternative model proposes that Rac inactivation generally rescues cytokinesis failure by reducing cortical tension, thus making it easier for the cell to divide when ring constriction is compromised. In this alternative model, CYK-4 was instead proposed to activate Rho by binding ECT-2. Using a combination of time-lapse in vivo single-cell analysis and Caenorhabditis elegans genetics, our evidence does not support this alternative model. First, we found that Rac disruption does not generally rescue cytokinesis failure: inhibition of Rac specifically rescues cytokinesis failure due to disruption of CYK-4 or ECT-2 but does not rescue cytokinesis failure due to disruption of two other contractile ring components, the Rho effectors diaphanous formin and myosin-II. Second, if CYK-4 regulates cytokinesis through Rho rather than Rac, then CYK-4 inhibition should decrease levels of downstream targets of Rho. Inconsistent with this, we found no change in the levels of f-actin or myosin-II at the division plane when CYK-4 GAP activity was reduced, suggesting that CYK-4 is not upstream of ECT-2/Rho activation. Instead, we found that the rescue of cytokinesis in CYK-4 mutants by Rac inactivation was Cdc42 dependent. Together our data suggest that CYK-4 GAP activity opposes Rac (and perhaps Cdc42) during cytokinesis.
Collapse
Affiliation(s)
- Yelena Zhuravlev
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Sophia M Hirsch
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Shawn N Jordan
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| | - Julien Dumont
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
43
|
Proteomics Screen Identifies Class I Rab11 Family Interacting Proteins as Key Regulators of Cytokinesis. Mol Cell Biol 2017; 37:MCB.00278-16. [PMID: 27872148 DOI: 10.1128/mcb.00278-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/11/2016] [Indexed: 01/08/2023] Open
Abstract
The 14-3-3 protein family orchestrates a complex network of molecular interactions that regulates various biological processes. Owing to their role in regulating the cell cycle and protein trafficking, 14-3-3 proteins are prevalent in human diseases such as cancer, diabetes, and neurodegeneration. 14-3-3 proteins are expressed in all eukaryotic cells, suggesting that they mediate their biological functions through evolutionarily conserved protein interactions. To identify these core 14-3-3 client proteins, we used an affinity-based proteomics approach to characterize and compare the human and Drosophila 14-3-3 interactomes. Using this approach, we identified a group of Rab11 effector proteins, termed class I Rab11 family interacting proteins (Rab11-FIPs), or Rip11 in Drosophila We found that 14-3-3 binds to Rip11 in a phospho-dependent manner to ensure its proper subcellular distribution during cell division. Our results indicate that Rip11 plays an essential role in the regulation of cytokinesis and that this function requires its association with 14-3-3 but not with Rab11. Together, our results suggest an evolutionarily conserved role for 14-3-3 in controlling Rip11-dependent protein transport during cytokinesis.
Collapse
|
44
|
Mishima M. Preparation of centralspindlin as an active heterotetramer of kinesin and GTPase activating protein subunits for in vitro structural and functional assays. Methods Cell Biol 2017; 137:371-385. [DOI: 10.1016/bs.mcb.2016.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
45
|
Kotýnková K, Su KC, West SC, Petronczki M. Plasma Membrane Association but Not Midzone Recruitment of RhoGEF ECT2 Is Essential for Cytokinesis. Cell Rep 2016; 17:2672-2686. [PMID: 27926870 PMCID: PMC5177604 DOI: 10.1016/j.celrep.2016.11.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 08/29/2016] [Accepted: 11/07/2016] [Indexed: 11/27/2022] Open
Abstract
Cytokinesis, the final step of cell division, begins with the formation of a cleavage furrow. How the mitotic spindle specifies the furrow at the equator in animal cells remains unknown. Current models propose that the concentration of the RhoGEF ECT2 at the spindle midzone and the equatorial plasma membrane directs furrow formation. Using chemical genetic and optogenetic tools, we demonstrate that the association of ECT2 with the plasma membrane during anaphase is required and sufficient for cytokinesis. Local membrane targeting of ECT2 leads to unilateral furrowing, highlighting the importance of local ECT2 activity. ECT2 mutations that prevent centralspindlin binding compromise concentration of ECT2 at the midzone and equatorial membrane but sustain cytokinesis. While the association of ECT2 with the plasma membrane is essential for cytokinesis, our data suggest that ECT2 recruitment to the spindle midzone is insufficient to account for equatorial furrowing and may act redundantly with yet-uncharacterized signals.
Collapse
Affiliation(s)
- Kristýna Kotýnková
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Herts EN6 3LD, UK; DNA Recombination and Repair Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kuan-Chung Su
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Herts EN6 3LD, UK; Whitehead Institute and Department of Biology, MIT, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Stephen C West
- DNA Recombination and Repair Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Mark Petronczki
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Herts EN6 3LD, UK; Boehringer Ingelheim RCV, Dr.-Boehringer-Gasse 5-11, 1121 Vienna, Austria.
| |
Collapse
|
46
|
Lawson CD, Der CJ. Filling GAPs in our knowledge: ARHGAP11A and RACGAP1 act as oncogenes in basal-like breast cancers. Small GTPases 2016; 9:290-296. [PMID: 27657701 DOI: 10.1080/21541248.2016.1220350] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Like RAS proteins, the aberrant function of RHO family small GTPases has been implicated in driving cancer development and growth. However, unlike the RAS family, where gain-of-function missense mutations are found in ∼25% of all human cancers, missense mutations are relatively rare in RHO proteins. Instead, altered RHO activity in cancer more commonly arises through the aberrant functions of RHO GTPase regulators. In many cancer types, altered expression and/or mutation of RHO-selective guanine nucleotide exchange factors (RHOGEFs) or GTPase-activating proteins (RHOGAPs), which activate or inactivate RHO GTPases, respectively, is observed. For example, deletion or loss of expression of the RHOA GAP DLC1 is well-established to drive cancer growth. Recently, we identified high expression of 2 RHOGAPs, ARHGAP11A and RACGAP1, in the basal-like breast cancer subtype. Unexpectedly, both of these RHOA GAPs exhibited properties of oncoproteins rather than tumor suppressors, in contrast to DLC1. In this commentary, we summarize our findings and speculate that different RHOA GAPs can play distinct roles in cancer depending on their spatial regulation and cancer type context. We also evaluate our results in light of recently-described cancer genome sequencing studies that have identified loss-of-function mutations of RHOA in specific cancer types.
Collapse
Affiliation(s)
- Campbell D Lawson
- a Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus , London , UK
| | - Channing J Der
- b Department of Pharmacology and Lineberger Comprehensive Cancer Center , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| |
Collapse
|
47
|
van Heesbeen RGHP, Raaijmakers JA, Tanenbaum ME, Halim VA, Lelieveld D, Lieftink C, Heck AJR, Egan DA, Medema RH. Aurora A, MCAK, and Kif18b promote Eg5-independent spindle formation. Chromosoma 2016; 126:473-486. [PMID: 27354041 PMCID: PMC5509784 DOI: 10.1007/s00412-016-0607-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 06/19/2016] [Accepted: 06/21/2016] [Indexed: 11/28/2022]
Abstract
Inhibition of the microtubule (MT) motor protein Eg5 results in a mitotic arrest due to the formation of monopolar spindles, making Eg5 an attractive target for anti-cancer therapies. However, Eg5-independent pathways for bipolar spindle formation exist, which might promote resistance to treatment with Eg5 inhibitors. To identify essential components for Eg5-independent bipolar spindle formation, we performed a genome-wide siRNA screen in Eg5-independent cells (EICs). We find that the kinase Aurora A and two kinesins, MCAK and Kif18b, are essential for bipolar spindle assembly in EICs and in cells with reduced Eg5 activity. Aurora A promotes bipolar spindle assembly by phosphorylating Kif15, hereby promoting Kif15 localization to the spindle. In turn, MCAK and Kif18b promote bipolar spindle assembly by destabilizing the astral MTs. One attractive way to interpret our data is that, in the absence of MCAK and Kif18b, excessive astral MTs generate inward pushing forces on centrosomes at the cortex that inhibit centrosome separation. Together, these data suggest a novel function for astral MTs in force generation on spindle poles and how proteins involved in regulating microtubule length can contribute to bipolar spindle assembly.
Collapse
Affiliation(s)
| | - Jonne A Raaijmakers
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marvin E Tanenbaum
- Hubrecht Institute, The Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Vincentius A Halim
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Daphne Lelieveld
- Cell Screening Core, Department of Cell Biology, Center for Molecular Medicine, University Medical Centre, Utrecht, The Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - David A Egan
- Cell Screening Core, Department of Cell Biology, Center for Molecular Medicine, University Medical Centre, Utrecht, The Netherlands
| | - René H Medema
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
48
|
Lawson CD, Fan C, Mitin N, Baker NM, George SD, Graham DM, Perou CM, Burridge K, Der CJ, Rossman KL. Rho GTPase Transcriptome Analysis Reveals Oncogenic Roles for Rho GTPase-Activating Proteins in Basal-like Breast Cancers. Cancer Res 2016; 76:3826-37. [PMID: 27216196 DOI: 10.1158/0008-5472.can-15-2923] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/18/2016] [Indexed: 02/06/2023]
Abstract
The basal-like breast cancer (BLBC) subtype accounts for a disproportionately high percentage of overall breast cancer mortality. The current therapeutic options for BLBC need improvement; hence, elucidating signaling pathways that drive BLBC growth may identify novel targets for the development of effective therapies. Rho GTPases have previously been implicated in promoting tumor cell proliferation and metastasis. These proteins are inactivated by Rho-selective GTPase-activating proteins (RhoGAP), which have generally been presumed to act as tumor suppressors. Surprisingly, RNA-Seq analysis of the Rho GTPase signaling transcriptome revealed high expression of several RhoGAP genes in BLBC tumors, raising the possibility that these genes may be oncogenic. To evaluate this, we examined the roles of two of these RhoGAPs, ArhGAP11A (also known as MP-GAP) and RacGAP1 (also known as MgcRacGAP), in promoting BLBC. Both proteins were highly expressed in human BLBC cell lines, and knockdown of either gene resulted in significant defects in the proliferation of these cells. Knockdown of ArhGAP11A caused CDKN1B/p27-mediated arrest in the G1 phase of the cell cycle, whereas depletion of RacGAP1 inhibited growth through the combined effects of cytokinesis failure, CDKN1A/p21-mediated RB1 inhibition, and the onset of senescence. Random migration was suppressed or enhanced by the knockdown of ArhGAP11A or RacGAP1, respectively. Cell spreading and levels of GTP-bound RhoA were increased upon depletion of either RhoGAP. We have established that, via the suppression of RhoA, ArhGAP11A and RacGAP1 are both critical drivers of BLBC growth, and propose that RhoGAPs can act as oncogenes in cancer. Cancer Res; 76(13); 3826-37. ©2016 AACR.
Collapse
Affiliation(s)
- Campbell D Lawson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Natalia Mitin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nicole M Baker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Samuel D George
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David M Graham
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Keith Burridge
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Channing J Der
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - Kent L Rossman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
49
|
Mishima M. Centralspindlin in Rappaport’s cleavage signaling. Semin Cell Dev Biol 2016; 53:45-56. [DOI: 10.1016/j.semcdb.2016.03.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 03/02/2016] [Indexed: 02/07/2023]
|
50
|
Henson JH, Buckley MW, Yeterian M, Weeks RM, Simerly CR, Shuster CB. Central Spindle Self-Organization and Cytokinesis in Artificially Activated Sea Urchin Eggs. THE BIOLOGICAL BULLETIN 2016; 230:85-95. [PMID: 27132131 DOI: 10.1086/bblv230n2p85] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The ability of microtubules of the mitotic apparatus to control the positioning and initiation of the cleavage furrow during cytokinesis was first established from studies on early echinoderm embryos. However, the identity of the microtubule population that imparts cytokinetic signaling is unclear. The two main--and not necessarily mutually exclusive--candidates are the central spindle and the astral rays. In the present study, we examined cytokinesis in ammonia-activated sea urchin eggs, which lack paternally derived centrosomes and undergo mitosis mediated by unusual anastral, bipolar mini-spindles. Live cell imaging and immunolabeling for microtubules and the centralspindlin constituent and kinesin-related protein, MKLP1, demonstrated that furrowing in ammonia-activated eggs was associated with aligned arrays of centralspindlin-linked, opposed bundles of antiparallel microtubules. These autonomous, zipper-like arrays were not associated with a mitotic apparatus, but did possess characteristics similar to the central spindle region of control, fertilized embryos. Our results highlight the self-organizing nature of the central spindle region and its ability to induce cytokinesis-like furrowing, even in the absence of a complete mitotic apparatus.
Collapse
Affiliation(s)
- John H Henson
- Department of Biology, Dickinson College, Carlisle, Pennsylvania 17013; Marine Biological Laboratory, Woods Hole, Massachusetts 02543;
| | - Mary W Buckley
- Department of Biology, Dickinson College, Carlisle, Pennsylvania 17013
| | - Mesrob Yeterian
- Department of Biology, Dickinson College, Carlisle, Pennsylvania 17013
| | - Richard M Weeks
- Department of Biology, Dickinson College, Carlisle, Pennsylvania 17013
| | - Calvin R Simerly
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213; and
| | - Charles B Shuster
- Marine Biological Laboratory, Woods Hole, Massachusetts 02543; Department of Biology, New Mexico State University, Las Cruces, New Mexico 88003
| |
Collapse
|