1
|
Jalali P, Samii A, Rezaee M, Shahmoradi A, Pashizeh F, Salehi Z. UBE2C: A pan-cancer diagnostic and prognostic biomarker revealed through bioinformatics analysis. Cancer Rep (Hoboken) 2024; 7:e2032. [PMID: 38577722 PMCID: PMC10995712 DOI: 10.1002/cnr2.2032] [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: 11/07/2023] [Revised: 02/03/2024] [Accepted: 02/22/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND The diverse and complex attributes of cancer have made it a daunting challenge to overcome globally and remains to endanger human life. Detection of critical cancer-related gene alterations in solid tumor samples better defines patient diagnosis and prognosis, and indicates what targeted therapies must be administered to improve cancer patients' outcome. MATERIALS AND METHODS To identify genes that have aberrant expression across different cancer types, differential expressed genes were detected within the TCGA datasets. Subsequently, the DEGs common to all pan cancers were determined. Furthermore, various methods were employed to gain genetic alterations, co-expression genes network and protein-protein interaction (PPI) network, pathway enrichment analysis of common genes. Finally, the gene regulatory network was constructed. RESULTS Intersectional analysis identified UBE2C as a common DEG between all 28 types of studied cancers. Upregulated UBE2C expression was significantly correlated with OS and DFS of 10 and 9 types of cancer patients. Also, UBE2C can be a diagnostic factor in CESC, CHOL, GBM, and UCS with AUC = 100% and diagnose 19 cancer types with AUC ≥90%. A ceRNA network constructed including UBE2C, 41 TFs, 10 shared miRNAs, and 21 circRNAs and 128 lncRNAs. CONCLUSION In summary, UBE2C can be a theranostic gene, which may serve as a reliable biomarker in diagnosing cancers, improving treatment responses and increasing the overall survival of cancer patients and can be a promising gene to be target by cancer drugs in the future.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical SciencesTehranIran
| | - Amir Samii
- Department of Hematology and Blood TransfusionSchool of Allied Medical Sciences, Iran University of Medical SciencesTehranIran
| | - Malihe Rezaee
- Department of PharmacologySchool of Medicine, Shahid Beheshti University of Medical SciencesTehranIran
| | - Arvin Shahmoradi
- Department of Laboratory MedicineFaculty of Paramedical, Kurdistan University of Medical SciencesSanandajIran
| | - Fatemeh Pashizeh
- Department of Clinical ImmunologyShahid Sadoughi University of Medical SciencesYazdIran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical SciencesTehranIran
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
2
|
Jing Y, Luo Y, Li L, Liu M, Liu JX. Deficiency of copper responsive gene stmn4 induces retinal developmental defects. Cell Biol Toxicol 2024; 40:2. [PMID: 38252267 PMCID: PMC10803583 DOI: 10.1007/s10565-024-09847-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
As part of the central nervous system (CNS), the retina senses light and also conducts and processes visual impulses. The damaged development of the retina not only causes visual damage, but also leads to epilepsy, dementia and other brain diseases. Recently, we have reported that copper (Cu) overload induces retinal developmental defects and down-regulates microtubule (MT) genes during zebrafish embryogenesis, but whether the down-regulation of microtubule genes mediates Cu stress induced retinal developmental defects is still unknown. In this study, we found that microtubule gene stmn4 exhibited obviously reduced expression in the retina of Cu overload embryos. Furthermore, stmn4 deficiency (stmn4-/-) resulted in retinal defects similar to those seen in Cu overload embryos, while overexpression of stmn4 effectively rescued retinal defects and cell apoptosis occurred in the Cu overload embryos and larvae. Meanwhile, stmn4 deficient embryos and larvae exhibited reduced mature retinal cells, the down-regulated expression of microtubules and cell cycle-related genes, and the mitotic cell cycle arrests of the retinal cells, which subsequently tended to apoptosis independent on p53. The results of this study demonstrate that Cu stress might lead to retinal developmental defects via down-regulating expression of microtubule gene stmn4, and stmn4 deficiency leads to impaired cell cycle and the accumulation of retinal progenitor cells (RPCs) and their subsequent apoptosis. The study provides a certain referee for copper overload in regulating the retinal development in fish.
Collapse
Affiliation(s)
- YuanYuan Jing
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yi Luo
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - LingYa Li
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
3
|
Yang J, Wang Z, Wang C, Tang D, Zang Z, Stover NA, Chen X, Li L. Single-cell transcriptome reveals cell division-regulated hub genes in the unicellular eukaryote Paramecium. Eur J Protistol 2023; 89:125978. [PMID: 37080141 DOI: 10.1016/j.ejop.2023.125978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/20/2023] [Accepted: 04/03/2023] [Indexed: 04/22/2023]
Abstract
The transition from growth to division during the cell cycle encompasses numerous conserved processes such as large-scale DNA replication and protein synthesis. In ciliate cells, asexual cell division is accompanied by additional cellular changes including amitotic nuclear division, extensive ciliogenesis, and trichocyst replication. However, the molecular mechanisms underlying these processes remain elusive. In this study, we present single-cell gene expression profiles of Paramecium cf. multimicronucleatum cells undergoing cell division. Our results reveal that the most up-regulated genes in dividing cells compared to growing cells are associated with 1) cell cycle signaling pathways including transcription, DNA replication, chromosome segregation and protein degradation; 2) microtubule proteins and tubulin glycylases which are essential for ciliogenesis, nuclei separation and structural differentiation signaling; and 3) trichocyst matrix proteins involved in trichocyst synthesis and reproduction. Furthermore, weighted gene co-expression network analysis identified hub genes that may play crucial roles during cell division. Our findings provide insights into cell cycle regulators, microtubules and trichocyst matrix proteins that may exert influence on this process in ciliates.
Collapse
Affiliation(s)
- Juan Yang
- Laboratory of Marine Protozoan Biodiversity & Evolution, Marine College, Shandong University, Weihai 264209, China
| | - Zhenyuan Wang
- Laboratory of Marine Protozoan Biodiversity & Evolution, Marine College, Shandong University, Weihai 264209, China
| | - Chundi Wang
- Laboratory of Marine Protozoan Biodiversity & Evolution, Marine College, Shandong University, Weihai 264209, China
| | - Danxu Tang
- Laboratory of Marine Protozoan Biodiversity & Evolution, Marine College, Shandong University, Weihai 264209, China
| | - Zihan Zang
- Laboratory of Marine Protozoan Biodiversity & Evolution, Marine College, Shandong University, Weihai 264209, China
| | - Naomi A Stover
- Department of Biology, Bradley University, Peoria 61625, USA
| | - Xiao Chen
- Laboratory of Marine Protozoan Biodiversity & Evolution, Marine College, Shandong University, Weihai 264209, China; Suzhou Research Institute, Shandong University, Suzhou 215123, China.
| | - Lifang Li
- Laboratory of Marine Protozoan Biodiversity & Evolution, Marine College, Shandong University, Weihai 264209, China.
| |
Collapse
|
4
|
Bahcivanci B, Shafiha R, Gkoutos GV, Acharjee A. Associating transcriptomics data with inflammatory markers to understand tumour microenvironment in hepatocellular carcinoma. Cancer Med 2023; 12:696-711. [PMID: 35715992 PMCID: PMC9844659 DOI: 10.1002/cam4.4941] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/25/2022] [Accepted: 06/03/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Liver cancer is the fourth leading cause of cancer-related death globally which is estimated to reach more than 1 million deaths a year by 2030. Among liver cancer types, hepatocellular carcinoma (HCC) accounts for approximately 90% of the cases and is known to have a tumour promoting inflammation regardless of its underlying aetiology. However, current promising treatment approaches, such as immunotherapy, are partially effective for most of the patients due to the immunosuppressive nature of the tumour microenvironment (TME). Therefore, there is an urgent need to fully understand TME in HCC and discover new immune markers to eliminate resistance to immunotherapy. METHODS We analyse three microarray datasets, using unsupervised and supervised methods, in an effort to discover signature genes. First, univariate, and multivariate, feature selection methods, such as the Boruta algorithm, are applied. Subsequently, an optimisation procedure, which utilises random forest algorithm with three dataset pairs combinations, is performed. The resulting optimal gene sets are then combined and further subjected to network analysis and pathway enrichment analysis so as to obtain information related to their biological relevance. The microarray datasets were analysed via the MCP-counter, CIBERSORT, TIMER, EPIC, and quanTIseq deconvolution methods and an estimation of cell type abundances for each dataset sample were identified. The differences in the cell type abundances, between the adjacent and tumour sample groups, were then assessed using a Wilcoxon Rank Sum test (p-value < 0.05). RESULTS The optimal gene signature sets, derived from each of the data pairs combination, achieved AUC values ranging from 0.959 to 0.988 in external validation sets using Random Forest model. CLEC1B and PTTG1 genes are retrieved across each optimal set. Among the signature genes, PTTG1, AURKA, and UBE2C genes are found to be involved in the regulation of mitotic sister chromatid separation and anaphase-promoting complex (APC) dependent catabolic process (adjusted p-value < 0.001). Additionally, the application of deconvolution algorithms revealed significant changes in cell type abundances of Regulatory T (Treg) cells, M0 and M1 macrophages, and T CD8+ cells between adjacent and tumour samples. CONCLUSION We identified ECM1 gene as a potential immune-related marker acting through immune cell migration and macrophage polarisation. Our results indicate that macrophages, such as M0 macrophage and M1 macrophage cells, undergo significant changes in HCC TME. Moreover, our immune deconvolution approach revealed significant infiltration of Treg cells and M0 macrophages, and a significant decrease in T CD8+ cells and M1 macrophages in tumour samples.
Collapse
Affiliation(s)
- Basak Bahcivanci
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational BiologyUniversity of BirminghamBirminghamUK
| | - Roshan Shafiha
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational BiologyUniversity of BirminghamBirminghamUK
| | - Georgios V. Gkoutos
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational BiologyUniversity of BirminghamBirminghamUK
- Institute of Translational MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
- NIHR Surgical Reconstruction and Microbiology Research CentreUniversity Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
- NIHR Experimental Cancer Medicine CentreBirminghamUK
| | - Animesh Acharjee
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational BiologyUniversity of BirminghamBirminghamUK
- Institute of Translational MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
- NIHR Surgical Reconstruction and Microbiology Research CentreUniversity Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
| |
Collapse
|
5
|
Circulating miRNA Expression Profiles and Machine Learning Models in Association with Response to Irinotecan-Based Treatment in Metastatic Colorectal Cancer. Int J Mol Sci 2022; 24:ijms24010046. [PMID: 36613487 PMCID: PMC9820223 DOI: 10.3390/ijms24010046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer represents a leading cause of cancer-related morbidity and mortality. Despite improvements, chemotherapy remains the backbone of colorectal cancer treatment. The aim of this study is to investigate the variation of circulating microRNA expression profiles and the response to irinotecan-based treatment in metastatic colorectal cancer and to identify relevant target genes and molecular functions. Serum samples from 95 metastatic colorectal cancer patients were analyzed. The microRNA expression was tested with a NucleoSpin miRNA kit (Machnery-Nagel, Germany), and a machine learning approach was subsequently applied for microRNA profiling. The top 10 upregulated microRNAs in the non-responders group were hsa-miR-181b-5p, hsa-miR-10b-5p, hsa-let-7f-5p, hsa-miR-181a-5p, hsa-miR-181d-5p, hsa-miR-301a-3p, hsa-miR-92a-3p, hsa-miR-155-5p, hsa-miR-30c-5p, and hsa-let-7i-5p. Similarly, the top 10 downregulated microRNAs were hsa-let-7d-5p, hsa-let-7c-5p, hsa-miR-215-5p, hsa-miR-143-3p, hsa-let-7a-5p, hsa-miR-10a-5p, hsa-miR-142-5p, hsa-miR-148a-3p, hsa-miR-122-5p, and hsa-miR-17-5p. The upregulation of microRNAs in the miR-181 family and the downregulation of those in the let-7 family appear to be mostly involved with non-responsiveness to irinotecan-based treatment.
Collapse
|
6
|
Yeo NJY, Wazny V, Nguyen NLU, Ng CY, Wu KX, Fan Q, Cheung CMG, Cheung C. Single-Cell Transcriptome of Wet AMD Patient-Derived Endothelial Cells in Angiogenic Sprouting. Int J Mol Sci 2022; 23:ijms232012549. [PMID: 36293401 PMCID: PMC9604336 DOI: 10.3390/ijms232012549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 12/01/2022] Open
Abstract
Age-related macular degeneration (AMD) is a global leading cause of visual impairment in older populations. ‘Wet’ AMD, the most common subtype of this disease, occurs when pathological angiogenesis infiltrates the subretinal space (choroidal neovascularization), causing hemorrhage and retinal damage. Gold standard anti-vascular endothelial growth factor (VEGF) treatment is an effective therapy, but the long-term prevention of visual decline has not been as successful. This warrants the need to elucidate potential VEGF-independent pathways. We generated blood out-growth endothelial cells (BOECs) from wet AMD and normal control subjects, then induced angiogenic sprouting of BOECs using a fibrin gel bead assay. To deconvolute endothelial heterogeneity, we performed single-cell transcriptomic analysis on the sprouting BOECs, revealing a spectrum of cell states. Our wet AMD BOECs share common pathways with choroidal neovascularization such as extracellular matrix remodeling that promoted proangiogenic phenotype, and our ‘activated’ BOEC subpopulation demonstrated proinflammatory hallmarks, resembling the tip-like cells in vivo. We uncovered new molecular insights that pathological angiogenesis in wet AMD BOECs could also be driven by interleukin signaling and amino acid metabolism. A web-based visualization of the sprouting BOEC single-cell transcriptome has been created to facilitate further discovery research.
Collapse
Affiliation(s)
- Natalie Jia Ying Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Vanessa Wazny
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Nhi Le Uyen Nguyen
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Chun-Yi Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Kan Xing Wu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Qiao Fan
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore 169857, Singapore
| | - Chui Ming Gemmy Cheung
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Singapore Eye Research Institute, Singapore 169856, Singapore
- Correspondence: (C.M.G.C.); (C.C.)
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore
- Correspondence: (C.M.G.C.); (C.C.)
| |
Collapse
|
7
|
Greil C, Engelhardt M, Wäsch R. The Role of the APC/C and Its Coactivators Cdh1 and Cdc20 in Cancer Development and Therapy. Front Genet 2022; 13:941565. [PMID: 35832196 PMCID: PMC9273091 DOI: 10.3389/fgene.2022.941565] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/08/2022] [Indexed: 12/03/2022] Open
Abstract
To sustain genomic stability by correct DNA replication and mitosis, cell cycle progression is tightly controlled by the cyclic activity of cyclin-dependent kinases, their binding to cyclins in the respective phase and the regulation of cyclin levels by ubiquitin-dependent proteolysis. The spindle assembly checkpoint plays an important role at the metaphase-anaphase transition to ensure a correct separation of sister chromatids before cytokinesis and to initiate mitotic exit, as an incorrect chromosome distribution may lead to genetically unstable cells and tumorigenesis. The ubiquitin ligase anaphase-promoting complex or cyclosome (APC/C) is essential for these processes by mediating the proteasomal destruction of cyclins and other important cell cycle regulators. To this end, it interacts with the two regulatory subunits Cdh1 and Cdc20. Both play a role in tumorigenesis with Cdh1 being a tumor suppressor and Cdc20 an oncogene. In this review, we summarize the current knowledge about the APC/C-regulators Cdh1 and Cdc20 in tumorigenesis and potential targeted therapeutic approaches.
Collapse
|
8
|
Vitovcova B, Skarkova V, Rudolf K, Rudolf E. Biology of Glioblastoma Multiforme-Exploration of Mitotic Catastrophe as a Potential Treatment Modality. Int J Mol Sci 2020; 21:ijms21155324. [PMID: 32727112 PMCID: PMC7432846 DOI: 10.3390/ijms21155324] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma multiforme (GBM) represents approximately 60% of all brain tumors in adults. This malignancy shows a high biological and genetic heterogeneity associated with exceptional aggressiveness, leading to a poor survival of patients. This review provides a summary of the basic biology of GBM cells with emphasis on cell cycle and cytoskeletal apparatus of these cells, in particular microtubules. Their involvement in the important oncosuppressive process called mitotic catastrophe will next be discussed along with select examples of microtubule-targeting agents, which are currently explored in this respect such as benzimidazole carbamate compounds. Select microtubule-targeting agents, in particular benzimidazole carbamates, induce G2/M cell cycle arrest and mitotic catastrophe in tumor cells including GBM, resulting in phenotypically variable cell fates such as mitotic death or mitotic slippage with subsequent cell demise or permanent arrest leading to senescence. Their effect is coupled with low toxicity in normal cells and not developed chemoresistance. Given the lack of efficient cytostatics or modern molecular target-specific compounds in the treatment of GBM, drugs inducing mitotic catastrophe might offer a new, efficient alternative to the existing clinical management of this at present incurable malignancy.
Collapse
|
9
|
Song Y, Song W, Li Z, Song W, Wen Y, Li J, Xia Q, Zhang M. CDC27 Promotes Tumor Progression and Affects PD-L1 Expression in T-Cell Lymphoblastic Lymphoma. Front Oncol 2020; 10:488. [PMID: 32391258 PMCID: PMC7190811 DOI: 10.3389/fonc.2020.00488] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
T-lymphoblastic lymphoma (T-LBL) is a rare hematological malignancy with highly aggressive, unique clinical manifestations, and poor prognosis. Cell division cycle 27 (CDC27) was previously reported to be a significant subunit of the anaphase-promoting complex/cyclosome. However, the specific functions and relevant mechanisms of CDC27 in T-LBL remain unknown. Through immunohistochemistry staining, we identified that CDC27 was overexpressed in T-LBL tissues and related to tumor progression and poor survival. Functional experiments demonstrated that CDC27 promoted proliferation in vivo and in vitro. Further experiment suggested the role of CDC27 in facilitating G1/S transition and promoting the expression of Cyclin D1 and CDK4. Then the effect of CDC27 in inhibiting apoptosis was also identified. Furthermore, we found a positive correlation between the expression of CDC27 and Programmed death ligand-1 (PD-L1) by immunohistochemistry staining. The interaction between CDC27 and PD-L1 was also proved by western blot, luciferase gene reporter assay and immunofluorescence. Taken together, our results showed that CDC27 contributes to T-LBL progression and there is a positive correlation between PD-L1 and CDC27, which offers novel perspectives for future studies on targeting CDC27 in T-LBL.
Collapse
Affiliation(s)
- Yue Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Academy of Medical Science of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Wei Song
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Academy of Medical Science of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Yibo Wen
- The Academy of Medical Science of Zhengzhou University, Zhengzhou, China
| | - Jiwei Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Qingxin Xia
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| |
Collapse
|
10
|
Wang L, Qu J, Liang Y, Zhao D, Rehman FU, Qin K, Zhang X. Identification and validation of key genes with prognostic value in non-small-cell lung cancer via integrated bioinformatics analysis. Thorac Cancer 2020; 11:851-866. [PMID: 32059076 PMCID: PMC7113067 DOI: 10.1111/1759-7714.13298] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 12/15/2022] Open
Abstract
Background Lung cancer is the most common cause of cancer‐related death among all human cancers and the five‐year survival rates are only 23%. The precise molecular mechanisms of non‐small cell lung cancer (NSCLC) are still unknown. The aim of this study was to identify and validate the key genes with prognostic value in lung tumorigenesis. Methods Four GEO datasets were obtained from the Gene Expression Omnibus (GEO) database. Common differentially expressed genes (DEGs) were selected for Kyoto Encyclopedia of Genes and Genomes pathway analysis and Gene Ontology enrichment analysis. Protein‐protein interaction (PPI) networks were constructed using the STRING database and visualized by Cytoscape software and Molecular Complex Detection (MCODE) were utilized to PPI network to pick out meaningful DEGs. Hub genes, filtered from the CytoHubba, were validated using the Gene Expression Profiling Interactive Analysis database. The expressions and prognostic values of hub genes were carried out through Gene Expression Profiling Interactive Analysis (GEPIA) and Kaplan‐Meier plotter. Finally, quantitative PCR and the Oncomine database were used to verify the differences in the expression of hub genes in lung cancer cells and tissues. Results A total of 121 DEGs (49 upregulated and 72 downregulated) were identified from four datasets. The PPI network was established with 121 nodes and 588 protein pairs. Finally, AURKA, KIAA0101, CDC20, MKI67, CHEK1, HJURP, and OIP5 were selected by Cytohubba, and they all correlated with worse overall survival (OS) in NSCLC. Conclusion The results showed that AURKA, KIAA0101, CDC20, MKI67, CHEK1, HJURP, and OIP5 may be critical genes in the development and prognosis of NSCLC. Key points Our results indicated that AURKA, KIAA0101, CDC20, MKI67, CHEK1, HJURP, and OIP5 may be critical genes in the development and prognosis of NSCLC. Our methods showed a new way to explore the key genes in cancer development.
Collapse
Affiliation(s)
- Li Wang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jialin Qu
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yu Liang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Deze Zhao
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Faisal Ul Rehman
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Kang Qin
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xiaochun Zhang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Wan WB, Wu K, Peng K, Qiu ZQ, Duan ZB, Chen X, Xu ZM, Cheng K, Zhao JMH, Shi QM. High level of RNF187 contributes to the progression and drug resistance of osteosarcoma. J Cancer 2020; 11:1351-1358. [PMID: 32047542 PMCID: PMC6995399 DOI: 10.7150/jca.33488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 10/05/2019] [Indexed: 01/20/2023] Open
Abstract
Objectives: Ring finger protein 187 (RNF187) was recently demonstrated to be up-regulation and function as a promoter in multiple cancers. However, the roles of RNF187 in osteosarcoma (OS) are unclear. Here, we tried to reveal the clinicopathological and biological roles of RNF187 in OS. Materials and Methods: We employed the quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) to determine the expression of RNF187 in OS tissues and cells. Migration and invasion capacities were analyzed by wound healing and transwell assays, and colony formation and CCK8 assays were performed to investigate proliferative ability. The functional effects of RNF187 on OS drugs resistance were further determined by CCK8 and western blot assays. Then, the relationship between RNF187 expression and clinical implications was analyzed by tissue microarrays (TMAs) including 51 OS cases. Moreover, the prognostic value was also determined by Kaplan-Meier analysis. Results: We reported that RNF187 mRNA was significantly increased in OS tissues compared to matched nontumorous tissues (3.83 ±0.79 vs. 1.70 ± 0.63), which was in line with the IHC assay in TMAs. By RNA interference and cDNA transfection, we showed high level of RNF187 increased the migration, invasion and proliferation of OS cells. Moreover, we demonstrated that elevated RNF187 expression induced OS cell drugs resistance, activated the ERK1/2 molecular and markedly enhanced the BCL-2 expression. Clinically, OS patients with high level of RNF187 was associated with Histologic differentiation (p=0.001), an advanced Enneking stage (p=0.001), response to chemotherapy (p=0.004), and metastasis (p= 0.001). Clinically, our data displayed that the RNF187 overexpression in OS samples associated with shorten overall survival (p=0.001) and high tumor recurrence (p=0.001) in postoperative OS patients. Conclusions: Our results indicate that Elevated RNF187 expression is a new adverse outcomes marker for OS patients and may be used as a new therapeutic target of OS.
Collapse
Affiliation(s)
- Wen-Bing Wan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Kai Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Kun Peng
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Zhi-Qiang Qiu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Zhi-Bin Duan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Xiang Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Ze-Min Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Ke Cheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Jiang-Ming-Hao Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Qing-Ming Shi
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| |
Collapse
|
12
|
Melloy PG. The anaphase-promoting complex: A key mitotic regulator associated with somatic mutations occurring in cancer. Genes Chromosomes Cancer 2019; 59:189-202. [PMID: 31652364 DOI: 10.1002/gcc.22820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase that helps control chromosome separation and exit from mitosis in many different kinds of organisms, including yeast, flies, worms, and humans. This review represents a new perspective on the connection between APC/C subunit mutations and cancer. The complex nature of APC/C and limited mutation analysis of its subunits has made it difficult to determine the relationship of each subunit to cancer. In this work, cancer genomic data were examined to identify APC/C subunits with a greater than 5% alteration frequency in 11 representative cancers using the cBioPortal database. Using the Genetic Determinants of Cancer Patient Survival database, APC/C subunits were also studied and found to be significantly associated with poor patient prognosis in several cases. In comparing these two kinds of cancer genomics data to published large-scale genomic analyses looking for cancer driver genes, ANAPC1 and ANAPC3/CDC27 stood out as being represented in all three types of analyses. Seven other subunits were found to be associated both with >5% alteration frequency in certain cancers and being associated with an effect on cancer patient prognosis. The aim of this review is to provide new approaches for investigators conducting in vivo studies of APC/C subunits and cancer progression. In turn, a better understanding of these APC/C subunits and their role in different cancers will help scientists design drugs that are more precisely targeted to certain cancers, using APC/C mutation status as a biomarker.
Collapse
Affiliation(s)
- Patricia G Melloy
- Department of Biological and Allied Health Sciences, Fairleigh Dickinson University, Madison, New Jersey
| |
Collapse
|
13
|
CDC20 expression in oestrogen receptor positive breast cancer predicts poor prognosis and lack of response to endocrine therapy. Breast Cancer Res Treat 2019; 178:535-544. [DOI: 10.1007/s10549-019-05420-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 08/22/2019] [Indexed: 12/23/2022]
|
14
|
Yang M, Jin Y, Fan S, Liang X, Jia J, Tan Z, Huang T, Li Y, Ma T, Li M. Inhibition of neddylation causes meiotic arrest in mouse oocyte. Cell Cycle 2019; 18:1254-1267. [PMID: 31111756 DOI: 10.1080/15384101.2019.1617453] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mammalian oocyte meiosis is a special form of cell division that provides haploid gametes for fertilization. Unlike in mitosis, post-translational modifications (PTMs) are more crucial during meiosis because of the absence of de novo mRNA transcription. As a classic PTM, protein neddylation is a biological process that mediates protein degradation by modifying cullin proteins and activating the Cullin-Ring E3 ligases. This process plays important roles in various biological processes such as autophagy and tumorigenesis. However, the function of neddylation in germ cells is unknown. In this study, we observed that the inhibition of neddylation by its specific inhibitor MLN4924 significantly arrests mouse oocyte at the stage of metaphase during meiosis. The arrested oocytes display impaired spindles with over-activation of spindle assembly checkpoint (SAC). Accordingly, we identified early mitosis inhibitor 1 (Emi1), a key inhibitor of anaphase-promoting complex/cyclosome (APC/CFzr1), as a substrate of neddylation-mediated protein degradation. Thus, our study uncovered an unknown role of neddylation in female germ cells and suggests that proper neddylation is essential for oocyte maturation.
Collapse
Affiliation(s)
- Mo Yang
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China.,b Key Laboratory of Assisted Reproduction , Ministry of Education , Beijing , China
| | - Yimei Jin
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China.,b Key Laboratory of Assisted Reproduction , Ministry of Education , Beijing , China
| | - Siying Fan
- c The Affiliated High School of Peking University , Beijing , China
| | - Xiaoling Liang
- d Department of Obstetrics and Gynecology , Peking University Shenzhen Hospital , Shenzhen , China
| | - Jialin Jia
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China.,b Key Laboratory of Assisted Reproduction , Ministry of Education , Beijing , China
| | - Zhongzhou Tan
- e School of Basic Medical Sciences , Peking University , Beijing , China
| | - Tao Huang
- e School of Basic Medical Sciences , Peking University , Beijing , China
| | - Yuan Li
- f Medical Center for Human Reproduction, Beijing Chaoyang Hospital , Capital Medical University , Beijing , China
| | - Teng Ma
- g Department of Cellular and Molecular Biology , Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute , Beijing , China.,h Beijing Key Laboratory for Radiobiology, Department of Radiation Toxicology and Oncology , Beijing Institute of Radiation Medicine , Beijing , China
| | - Mo Li
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China.,b Key Laboratory of Assisted Reproduction , Ministry of Education , Beijing , China
| |
Collapse
|
15
|
Ewerth D, Kreutmair S, Schmidts A, Ihorst G, Follo M, Wider D, Felthaus J, Schüler J, Duyster J, Illert AL, Engelhardt M, Wäsch R. APC/C Cdh1 regulates the balance between maintenance and differentiation of hematopoietic stem and progenitor cells. Cell Mol Life Sci 2019; 76:369-380. [PMID: 30357422 PMCID: PMC11105657 DOI: 10.1007/s00018-018-2952-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/13/2018] [Accepted: 10/15/2018] [Indexed: 10/28/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) represent the lifelong source of all blood cells and continuously regenerate the hematopoietic system through differentiation and self-renewal. The process of differentiation is initiated in the G1 phase of the cell cycle, when stem cells leave their quiescent state. During G1, the anaphase-promoting complex or cyclosome associated with the coactivator Cdh1 is highly active and marks proteins for proteasomal degradation to regulate cell proliferation. Following Cdh1 knockdown in HSPCs, we analyzed human and mouse hematopoiesis in vitro and in vivo in competitive transplantation assays. We found that Cdh1 is highly expressed in human CD34+ HSPCs and downregulated in differentiated subsets; whereas, loss of Cdh1 restricts myeloid differentiation, supports B cell development and preserves immature short-term HSPCs without affecting proliferation or viability. Our data highlight a role of Cdh1 as a regulator of balancing the maintenance of HSPCs and differentiation into mature blood cells.
Collapse
Affiliation(s)
- Daniel Ewerth
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, 79104, Freiburg, Germany
| | - Stefanie Kreutmair
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Andrea Schmidts
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Gabriele Ihorst
- Clinical Trials Unit, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Marie Follo
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Dagmar Wider
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Julia Felthaus
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | | | - Justus Duyster
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Lena Illert
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Monika Engelhardt
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Ralph Wäsch
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Hugstetter Strasse 55, 79106, Freiburg, Germany.
| |
Collapse
|
16
|
Harkness TAA. Activating the Anaphase Promoting Complex to Enhance Genomic Stability and Prolong Lifespan. Int J Mol Sci 2018; 19:ijms19071888. [PMID: 29954095 PMCID: PMC6073722 DOI: 10.3390/ijms19071888] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 12/19/2022] Open
Abstract
In aging cells, genomic instability is now recognized as a hallmark event. Throughout life, cells encounter multiple endogenous and exogenous DNA damaging events that are mostly repaired, but inevitably DNA mutations, chromosome rearrangements, and epigenetic deregulation begins to mount. Now that people are living longer, more and more late life time is spent suffering from age-related disease, in which genomic instability plays a critical role. However, several major questions remain heavily debated, such as the following: When does aging start? How long can we live? In order to minimize the impact of genomic instability on longevity, it is important to understand when aging starts, and to ensure repair mechanisms remain optimal from the very start to the very end. In this review, the interplay between the stress and nutrient response networks, and the regulation of homeostasis and genomic stability, is discussed. Mechanisms that link these two networks are predicted to be key lifespan determinants. The Anaphase Promoting Complex (APC), a large evolutionarily conserved ubiquitin ligase, can potentially serve this need. Recent work demonstrates that the APC maintains genomic stability, mounts a stress response, and increases longevity in yeast. Furthermore, inhibition of APC activity by glucose and nutrient response factors indicates a tight link between the APC and the stress/nutrient response networks.
Collapse
Affiliation(s)
- Troy A A Harkness
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
17
|
Nayak D, Sivaraman J. Structure of LNX1:Ubc13~Ubiquitin Complex Reveals the Role of Additional Motifs for the E3 Ligase Activity of LNX1. J Mol Biol 2018; 430:1173-1188. [PMID: 29496391 DOI: 10.1016/j.jmb.2018.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 02/17/2018] [Accepted: 02/19/2018] [Indexed: 10/17/2022]
Abstract
LNX1 (ligand of numb protein-X1) is a RING and PDZ domain-containing E3 ubiquitin ligase that ubiquitinates human c-Src kinase. Here, we report the identification and structure of the ubiquitination domain of LNX1, the identification of Ubc13/Ube2V2 as a functional E2 in vitro, and the structural and functional studies of the Ubc13~Ub intermediate in complex with the ubiquitination domain of LNX1. The RING domain of LNX1 is embedded between two zinc-finger motifs (Zn-RING-Zn), both of which are crucial for its ubiquitination activity. In the heterodimeric complex, the ubiquitin of one monomer shares more buried surface area with LNX1 of the other monomer and these interactions are unique and essential for catalysis. This study reveals how the LNX1 RING domain is structurally and mechanistically dependent on other motifs for its E3 ligase activity, and describes how dimeric LNX1 recruits ubiquitin-loaded Ubc13 for Ub transfer via E3 ligase-mediated catalysis.
Collapse
Affiliation(s)
- Digant Nayak
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - J Sivaraman
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543.
| |
Collapse
|
18
|
Xiong D, Zhu SQ, Wu YB, Jin C, Jiang JH, Liao YF, Long X, Wu HB, Xu JJ, Li JJ, Ding JY. Ring finger protein 38 promote non-small cell lung cancer progression by endowing cell EMT phenotype. J Cancer 2018; 9:841-850. [PMID: 29581762 PMCID: PMC5868148 DOI: 10.7150/jca.23138] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/29/2017] [Indexed: 01/11/2023] Open
Abstract
Objectives: Ring finger protein 38 (RNF38), as an E3 ubiquitin ligase, plays an essential role in multiple biological processes by controlling cell apoptosis, cell cycle and DNA repair, and resides in chromosome 9 (9p13) which is involvement in cancer pathogenesis including lung cancer. However, its function in tumorigenesis remains unclear. Hence, this study set out to investigate the biological function and clinical implications of RNF38 in non-small cell lung cancer (NSCLC). Materials and Methods: Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were used to detect RNF38 protein and mRNA levels in NSCLC and corresponding paratumor tissues. Tissue microarrays (TMA) analysis of 208 NSCLC cases were used to evaluate the relationship between RNF38 expression and clinical implications. Prognostic value was assessed by Kaplan-Meier analysis and log-rank tests. Wound-healing assays, trans-well assays, colony formation assays and CCK8 were used to assess cell migration, invasion and proliferative ability respectively. The analysis of epithelial-to-mesenchymal transition (EMT) phenotype was carried out by immunofluorescence and western blot. Results: Our data revealed that elevated RNF38 expression were more common in NSCLC tissues than paired normal tissues in both mRNA (2.82 ± 0.29 vs. 1.23 ± 0.13) and protein (2.75 ± 0.09 vs. 1.24 ± 0.02) level. High levels of RNF38 expression were significantly associated with lymph node metastases, higher TNM stages (p=0.011), larger tumor size (p=2.09E-04) and predicted poor prognosis. RNF38 expression was inversely correlated with E-cadherin expression (P= 0.025). Moreover, downregulation of RNF38 impaired the proliferation, metastatic and invasive abilities in NSCLC cells. In addition, aberrant RNF38 expression could modulate the key molecules of EMT. Conclusions: Our results indicate that elevated expression of RNF38 is significantly associated with the proliferation and metastatic capacity of NSCLC cells, and RNF38 overexpression can serve as a biomarker of NSCLC poor prognosis.
Collapse
Affiliation(s)
- Dian Xiong
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, P. R. China.,Department of Thoracic Surgery, The Affiliated Zhongshan Hospital of Fudan University, Shanghai 200032, P. R. China
| | - Shu-Qiang Zhu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, P. R. China
| | - Yong-Bing Wu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, P. R. China
| | - Chun Jin
- Department of Thoracic Surgery, The Affiliated Zhongshan Hospital of Fudan University, Shanghai 200032, P. R. China
| | - Jia-Hao Jiang
- Department of Thoracic Surgery, The Affiliated Zhongshan Hospital of Fudan University, Shanghai 200032, P. R. China
| | - Yun-Fei Liao
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, P. R. China
| | - Xiang Long
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, P. R. China
| | - Hai-Bo Wu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, P. R. China
| | - Jian-Jun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, P. R. China
| | - Ji-Jun Li
- Department of Cardio-Thoracic Surgery, Kashgar Prefecture Second People's Hospital, Kashgar, Xinjiang 844000, China
| | - Jian-Yong Ding
- Department of Thoracic Surgery, The Affiliated Zhongshan Hospital of Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
19
|
Melanoma antigen-D2 controls cell cycle progression and modulates the DNA damage response. Biochem Pharmacol 2018; 153:217-229. [PMID: 29371029 DOI: 10.1016/j.bcp.2018.01.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/19/2018] [Indexed: 01/28/2023]
Abstract
Overexpression of the ubiquitous type II melanoma antigen-D2 (MAGED2) in numerous types of cancer suggests that this protein contributes to carcinogenesis, a well-documented characteristic of other MAGE proteins. Modification of MAGED2 intracellular localization during cell cycle phases and following treatment with camptothecin (CPT) and phosphorylation by ATM/ATR following ionizing irradiation led us to investigate the molecular functions of MAGED2 in the cellular response to DNA damage. Cell cycle regulators, cell cycle progression, and bromodeoxyuridine (BrdU) incorporation were compared between MAGED2-sufficient and -depleted U2OS cells following exposure to CPT. At 24 h post-CPT removal, MAGED2-depleted cells had lower levels of p21 and p27, and there was an increase in S phase BrdU-positive cells with a concurrent decrease in cells in G2. These cell cycle modifications were p21-independent, but ATR-, SKP2-, and CDC20-dependent. Importantly, while MAGED2 depletion reduced CHK2 phosphorylation after 8 h of CPT treatment, it enhanced and prolonged CHK1 phosphorylation after a 24 h recovery period, indicating sustained ATR activation. MAGED2 depletion had no impact on cell survival under our experimental conditions. In summary, our data indicate that MAGED2 reduced CPT-related replicative stress, suggesting a role for this protein in genomic stability.
Collapse
|
20
|
Schnerch D, Schüler J, Follo M, Felthaus J, Wider D, Klingner K, Greil C, Duyster J, Engelhardt M, Wäsch R. Proteasome inhibition enhances the efficacy of volasertib-induced mitotic arrest in AML in vitro and prolongs survival in vivo. Oncotarget 2017; 8:21153-21166. [PMID: 28416751 PMCID: PMC5400573 DOI: 10.18632/oncotarget.15503] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/07/2017] [Indexed: 11/25/2022] Open
Abstract
Elderly and frail patients, diagnosed with acute myeloid leukemia (AML) and ineligible to undergo intensive treatment, have a dismal prognosis. The small molecule inhibitor volasertib induces a mitotic block via inhibition of polo-like kinase 1 and has shown remarkable anti-leukemic activity when combined with low-dose cytarabine. We have demonstrated that AML cells are highly vulnerable to cell death in mitosis yet manage to escape a mitotic block through mitotic slippage by sustained proteasome-dependent slow degradation of cyclin B. Therefore, we tested whether interfering with mitotic slippage through proteasome inhibition arrests and kills AML cells more efficiently during mitosis. We show that therapeutic doses of bortezomib block the slow degradation of cyclin B during a volasertib-induced mitotic arrest in AML cell lines and patient-derived primary AML cells. In a xenotransplant mouse model of human AML, mice receiving volasertib in combination with bortezomib showed superior disease control compared to mice receiving volasertib alone, highlighting the potential therapeutic impact of this drug combination.
Collapse
Affiliation(s)
- Dominik Schnerch
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Marie Follo
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Felthaus
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dagmar Wider
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Christine Greil
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Justus Duyster
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Monika Engelhardt
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ralph Wäsch
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
21
|
|
22
|
Frequent mutations in acetylation and ubiquitination sites suggest novel driver mechanisms of cancer. Genome Med 2016; 8:55. [PMID: 27175787 PMCID: PMC4864925 DOI: 10.1186/s13073-016-0311-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 04/19/2016] [Indexed: 12/14/2022] Open
Abstract
Background Discovery of cancer drivers is a major goal of cancer research. Driver genes and pathways are often predicted using mutation frequency, assuming that statistically significant recurrence of specific somatic mutations across independent samples indicates their importance in cancer. However, many mutations, including known cancer drivers, are not observed at high frequency. Fortunately, abundant information is available about functional “active sites” in proteins that can be integrated with mutations to predict cancer driver genes, even based on low frequency mutations. Further, considering active site information predicts detailed biochemical mechanisms impacted by the mutations. Post-translational modifications (PTMs) are active sites that are regulatory switches in proteins and pathways. We analyzed acetylation and ubiquitination, two important PTM types often involved in chromatin organization and protein degradation, to find proteins that are significantly affected by tumor somatic mutations. Methods We performed computational analyses of acetylation and ubiquitination sites in a pan-cancer dataset of 3200 tumor samples from The Cancer Genome Atlas (TCGA). These analyses were targeted at different levels of biological organization including individual genes, pathway annotated gene sets, and protein-protein interaction networks. Results Acetylation and ubiquitination site mutations are enriched in cancer with significantly stronger evolutionary conservation and accumulation in protein domains. Gene-focused analysis with the ActiveDriver method reveals significant co-occurrences of acetylation and ubiquitination PTMs and mutation hotspots in known oncoproteins (TP53, AKT1, IDH1) and highlights candidate cancer driver genes with PTM-related mechanisms (e.g. several histone proteins and the splicing factor SF3B1). Pathway analysis shows that PTM mutations in acetylation and ubiquitination sites accumulate in cancer-related processes such as cell cycle, apoptosis, chromatin regulation, and metabolism. Integrated mutation analysis of clinical information and protein interaction networks suggests that many PTM-specific mutations associate with decreased patient survival. Conclusions Mutation analysis of acetylation and ubiquitination PTM sites reveals their importance in cancer. As PTM networks are increasingly mapped and related enzymes are often druggable, deeper investigation of specific associated mutations may lead to the discovery of treatment-relevant cellular mechanisms. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0311-2) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Sato-Kuwabara Y, Fregnani JHTG, Jampietro J, Carvalho KC, Franco CP, da Costa WL, Coimbra FJF, Soares FA. Comparative analysis of basaloid and conventional squamous cell carcinomas of the esophagus: prognostic relevance of clinicopathological features and protein expression. Tumour Biol 2015; 37:6691-9. [PMID: 26649862 DOI: 10.1007/s13277-015-4551-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/30/2015] [Indexed: 12/01/2022] Open
Abstract
Basaloid squamous cell carcinoma (BSCC), a variant of squamous cell carcinoma (SCC), is a rare and aggressive epithelial malignancy which has been reported in only 0.1-11 % of primary esophageal carcinomas. In this study, a comparison of clinicopathological features and protein expression between esophageal BSCC (EBSCC) and conventional esophageal SCC (ESCC) cases from Brazil was performed in order to find factors that can be relevant to better characterize EBSCC. The expression of HER2, epidermal growth factor receptor (EGFR), Ki-67, and cyclins (A, B1, and D1) in 111 cases (95 ESCC and 16 EBSCC) was evaluated by immunohistochemistry using tissue microarray. When the clinicopathological data were compared, no significant difference was found between the two histological types. Although the difference is not significant (p = 0.055), the EGFR expression was more frequent in the conventional ESCC than in the EBSCC group. Our results indicate that the clinicopathological profiles of conventional ESCC and EBSCC are similar and provide no indicators for differences in prognosis between these two groups.
Collapse
Affiliation(s)
- Yukie Sato-Kuwabara
- International Research Center, A.C. Camargo Cancer Center, Rua Taguá, 440 Liberdade, São Paulo, SP, 01508-010, Brazil.
| | - José Humberto T G Fregnani
- Gynecologic Oncology Department, Fundação Pio XII, Hospital do Câncer de Barretos, Rua Antenor Duarte Villela, 1331, Barretos, SP, 14784-400, Brazil
| | - Juliano Jampietro
- International Research Center, A.C. Camargo Cancer Center, Rua Taguá, 440 Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Katia Cândido Carvalho
- Obstetrics and Gynecology Department, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, São Paulo, SP, 01246-903, Brazil
| | - Carolina Parucce Franco
- Department of Abdominal Surgery, A.C. Camargo Cancer Center, Rua Antônio Prudente, 211, 1o. andar, São Paulo, SP, 01509-010, Brazil
| | - Wilson Luís da Costa
- Department of Abdominal Surgery, A.C. Camargo Cancer Center, Rua Antônio Prudente, 211, 1o. andar, São Paulo, SP, 01509-010, Brazil
| | - Felipe J F Coimbra
- Department of Abdominal Surgery, A.C. Camargo Cancer Center, Rua Antônio Prudente, 211, 1o. andar, São Paulo, SP, 01509-010, Brazil
| | - Fernando Augusto Soares
- Department of Anatomic Pathology, A.C. Camargo Cancer Center, Rua Antônio Prudente, 211, 1o. andar, São Paulo, SP, 01509-010, Brazil.,Department of Stomatology, School of Dentistry, University of São Paulo, Av. Professor Lineu Prestes, 2227, São Paulo, SP, 05508-000, Brazil
| |
Collapse
|
24
|
The role of APC/C(Cdh1) in replication stress and origin of genomic instability. Oncogene 2015; 35:3062-70. [PMID: 26455319 DOI: 10.1038/onc.2015.367] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 01/01/2023]
Abstract
It has been proposed that the APC/C(Cdh1) functions as a tumor suppressor by maintaining genomic stability. However, the exact nature of genomic instability following loss of Cdh1 is unclear. Using biochemistry and live cell imaging of single cells we found that Cdh1 knockdown (kd) leads to strong nuclear stabilization of the substrates cyclin A and B and deregulated kinetics of DNA replication. Restoration of the Cdh1-dependent G2 DNA damage checkpoint did not result in G2 arrest but blocked cells in prometaphase, suggesting that these cells enter mitosis despite incomplete replication. This results in DNA double-strand breaks, anaphase bridges, cytokinesis defects and tetraploidization. Tetraploid cells are the source of supernumerary centrosomes following Cdh1-kd, leading to multipolar mitosis or centrosome clustering, in turn resulting in merotelic attachment and lagging chromosomes. Whereas some of these events cause apoptosis during mitosis, surviving cells may accumulate chromosomal aberrations.
Collapse
|
25
|
Impaired Cell Cycle Regulation in a Natural Equine Model of Asthma. PLoS One 2015; 10:e0136103. [PMID: 26292153 PMCID: PMC4546272 DOI: 10.1371/journal.pone.0136103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 07/29/2015] [Indexed: 12/14/2022] Open
Abstract
Recurrent airway obstruction (RAO) is a common and potentially debilitating lower airway disease in horses, which shares many similarities with human asthma. In susceptible horses RAO exacerbation is caused by environmental allergens and irritants present in hay dust. The objective of this study was the identification of genes and pathways involved in the pathology of RAO by global transcriptome analyses in stimulated peripheral blood mononuclear cells (PBMCs). We performed RNA-seq on PBMCs derived from 40 RAO affected and 45 control horses belonging to three cohorts of Warmblood horses: two half-sib families and one group of unrelated horses. PBMCs were stimulated with hay dust extract, lipopolysaccharides, a recombinant parasite antigen, or left unstimulated. The total dataset consisted of 561 individual samples. We detected significant differences in the expression profiles between RAO and control horses. Differential expression (DE) was most marked upon stimulation with hay dust extract. An important novel finding was a strong upregulation of CXCL13 together with many genes involved in cell cycle regulation in stimulated samples from RAO affected horses, in addition to changes in the expression of several HIF-1 transcription factor target genes. The RAO condition alters systemic changes observed as differential expression profiles of PBMCs. Those changes also depended on the cohort and stimulation of the samples and were dominated by genes involved in immune cell trafficking, development, and cell cycle regulation. Our findings indicate an important role of CXCL13, likely macrophage or Th17 derived, and the cell cycle regulator CDC20 in the immune response in RAO.
Collapse
|
26
|
Gayyed MF, El-Maqsoud NMRA, Tawfiek ER, El Gelany SAA, Rahman MFA. A comprehensive analysis of CDC20 overexpression in common malignant tumors from multiple organs: its correlation with tumor grade and stage. Tumour Biol 2015; 37:749-62. [DOI: 10.1007/s13277-015-3808-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/20/2015] [Indexed: 11/29/2022] Open
|
27
|
Moreno SP, Gambus A. Regulation of Unperturbed DNA Replication by Ubiquitylation. Genes (Basel) 2015; 6:451-68. [PMID: 26121093 PMCID: PMC4584310 DOI: 10.3390/genes6030451] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/05/2015] [Accepted: 06/16/2015] [Indexed: 02/07/2023] Open
Abstract
Posttranslational modification of proteins by means of attachment of a small globular protein ubiquitin (i.e., ubiquitylation) represents one of the most abundant and versatile mechanisms of protein regulation employed by eukaryotic cells. Ubiquitylation influences almost every cellular process and its key role in coordination of the DNA damage response is well established. In this review we focus, however, on the ways ubiquitylation controls the process of unperturbed DNA replication. We summarise the accumulated knowledge showing the leading role of ubiquitin driven protein degradation in setting up conditions favourable for replication origin licensing and S-phase entry. Importantly, we also present the emerging major role of ubiquitylation in coordination of the active DNA replication process: preventing re-replication, regulating the progression of DNA replication forks, chromatin re-establishment and disassembly of the replisome at the termination of replication forks.
Collapse
Affiliation(s)
- Sara Priego Moreno
- School of Cancer Sciences, University of Birmingham, Vincent Drive, B15 2TT, Birmingham, UK
| | - Agnieszka Gambus
- School of Cancer Sciences, University of Birmingham, Vincent Drive, B15 2TT, Birmingham, UK.
| |
Collapse
|
28
|
Lafranchi L, de Boer HR, de Vries EGE, Ong SE, Sartori AA, van Vugt MATM. APC/C(Cdh1) controls CtIP stability during the cell cycle and in response to DNA damage. EMBO J 2014; 33:2860-79. [PMID: 25349192 DOI: 10.15252/embj.201489017] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human cells have evolved elaborate mechanisms for responding to DNA damage to maintain genome stability and prevent carcinogenesis. For instance, the cell cycle can be arrested at different stages to allow time for DNA repair. The APC/C(C) (dh1) ubiquitin ligase mainly regulates mitotic exit but is also implicated in the DNA damage-induced G2 arrest. However, it is currently unknown whether APC/C(C) (dh1) also contributes to DNA repair. Here, we show that Cdh1 depletion causes increased levels of genomic instability and enhanced sensitivity to DNA-damaging agents. Using an integrated proteomics and bioinformatics approach, we identify CtIP, a DNA-end resection factor, as a novel APC/C(C) (dh1) target. CtIP interacts with Cdh1 through a conserved KEN box, mutation of which impedes ubiquitylation and downregulation of CtIP both during G1 and after DNA damage in G2. Finally, we find that abrogating the CtIP-Cdh1 interaction results in delayed CtIP clearance from DNA damage foci, increased DNA-end resection, and reduced homologous recombination efficiency. Combined, our results highlight the impact of APC/C(C) (dh1) on the maintenance of genome integrity and show that this is, at least partially, achieved by controlling CtIP stability in a cell cycle- and DNA damage-dependent manner.
Collapse
Affiliation(s)
- Lorenzo Lafranchi
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Harmen R de Boer
- Department of Medical Oncology, University Medical Center Groningen University of Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen University of Groningen, Groningen, The Netherlands
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Alessandro A Sartori
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Marcel A T M van Vugt
- Department of Medical Oncology, University Medical Center Groningen University of Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Ayuda-Durán P, Devesa F, Gomes F, Sequeira-Mendes J, Avila-Zarza C, Gómez M, Calzada A. The CDK regulators Cdh1 and Sic1 promote efficient usage of DNA replication origins to prevent chromosomal instability at a chromosome arm. Nucleic Acids Res 2014; 42:7057-68. [PMID: 24753426 PMCID: PMC4066753 DOI: 10.1093/nar/gku313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Robustness and completion of DNA replication rely on redundant DNA replication origins. Reduced efficiency of origin licensing is proposed to contribute to chromosome instability in CDK-deregulated cell cycles, a frequent alteration in oncogenesis. However, the mechanism by which this instability occurs is largely unknown. Current models suggest that limited origin numbers would reduce fork density favouring chromosome rearrangements, but experimental support in CDK-deregulated cells is lacking. We have investigated the pattern of origin firing efficiency in budding yeast cells lacking the CDK regulators Cdh1 and Sic1. We show that each regulator is required for efficient origin activity, and that both cooperate non-redundantly. Notably, origins are differentially sensitive to CDK deregulation. Origin sensitivity is independent on normal origin efficiency, firing timing or chromosomal location. Interestingly, at a chromosome arm, there is a shortage of origin firing involving active and dormant origins, and the extent of shortage correlates with the severity of CDK deregulation and chromosome instability. We therefore propose that CDK deregulation in G1 phase compromises origin redundancy by decreasing the number of active and dormant origins, leading to origin shortage and increased chromosome instability.
Collapse
Affiliation(s)
- Pilar Ayuda-Durán
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología CNB-CSIC, Darwin 3, Madrid 28049, Spain
| | - Fernando Devesa
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología CNB-CSIC, Darwin 3, Madrid 28049, Spain
| | - Fábia Gomes
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología CNB-CSIC, Darwin 3, Madrid 28049, Spain
| | - Joana Sequeira-Mendes
- Centro de Biología Molecular Severo Ochoa CBMSO-CSIC/UAM, Nicolás Cabrera 1, Madrid 28049, Spain
| | | | - María Gómez
- Centro de Biología Molecular Severo Ochoa CBMSO-CSIC/UAM, Nicolás Cabrera 1, Madrid 28049, Spain
| | - Arturo Calzada
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología CNB-CSIC, Darwin 3, Madrid 28049, Spain
| |
Collapse
|
30
|
Dou QP. Deubiquitinating Enzymes as Novel Targets for Cancer Therapies. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2014. [PMCID: PMC7123001 DOI: 10.1007/978-3-319-06752-0_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Most ubiquitinated proteins can be recognized and degraded by the 26S proteasome. In the meantime, protein deubiquitination by various deubiquitinating enzymes (DUBs) regulates protein stability within cells, and it can counterbalance intracellular homeostasis mediated by ubiquitination. Numerous reports have demonstrated that an aberrant process of the ubiquitin-proteasome pathway (UPP) regulated by the ubiquitination and deubiquitination systems results in failure of balancing between protein stability and degradation, and this failure can lead to tumorigenesis in various organs and tissues of mammals. The identification of molecular properties for various DUBs is very critical to understand cancer development and tumorigenesis. Therefore, knowledge of DUBs and their association with cancer and diseases is indispensible for developing effective inhibitors for DUBs. This chapter describes various features and functions of cancer-related DUBs. In addition, we summarize several inhibitors that specifically target certain DUBs in cancer and suggest that DUBs may be one of the most ideal and attractive therapeutic targets.
Collapse
Affiliation(s)
- Q. Ping Dou
- Wayne State University, Detroit, Michigan USA
| |
Collapse
|
31
|
Chen YS, Qiu XB. Ubiquitin at the crossroad of cell death and survival. CHINESE JOURNAL OF CANCER 2013; 32:640-7. [PMID: 23816559 PMCID: PMC3870847 DOI: 10.5732/cjc.012.10283] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 02/20/2013] [Accepted: 03/15/2013] [Indexed: 12/19/2022]
Abstract
Ubiquitination is crucial for cellular processes, such as protein degradation, apoptosis, autophagy, and cell cycle progression. Dysregulation of the ubiquitination network accounts for the development of numerous diseases, including cancer. Thus, targeting ubiquitination is a promising strategy in cancer therapy. Both apoptosis and autophagy are involved in tumorigenesis and response to cancer therapy. Although both are categorized as types of cell death, autophagy is generally considered to have protective functions, including protecting cells from apoptosis under certain cellular stress conditions. This review highlights recent advances in understanding the regulation of apoptosis and autophagy by ubiquitination.
Collapse
Affiliation(s)
- Yu-Shan Chen
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, and College of Life Sciences, Beijing Normal University, Beijing 100875, P. R. China.
| | | |
Collapse
|
32
|
Lu L, Hu S, Wei R, Qiu X, Lu K, Fu Y, Li H, Xing G, Li D, Peng R, He F, Zhang L. The HECT type ubiquitin ligase NEDL2 is degraded by anaphase-promoting complex/cyclosome (APC/C)-Cdh1, and its tight regulation maintains the metaphase to anaphase transition. J Biol Chem 2013; 288:35637-50. [PMID: 24163370 DOI: 10.1074/jbc.m113.472076] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
NEDD4-like ubiquitin ligase 2 (NEDL2) is a HECT type ubiquitin ligase. NEDL2 enhances p73 transcriptional activity and degrades ATR kinase in lamin misexpressed cells. Compared with the important functions of other HECT type ubiquitin ligase, there is less study concerning the function and regulation of NEDL2. Using primary antibody immunoprecipitation and mass spectrometry, we identify a list of potential proteins that are putative NEDL2-interacting proteins. The candidate list contains many of mitotic proteins, especially including several subunits of anaphase-promoting complex/cyclosome (APC/C) and Cdh1, an activator of APC/C. Cdh1 can interact with NEDL2 in vivo and in vitro. Cdh1 recognizes one of the NEDL2 destruction boxes (R(740)GSL(743)) and targets it for degradation in an APC/C-dependent manner during mitotic exit. Overexpression of Cdh1 reduces the protein level of NEDL2, whereas knockdown of Cdh1 increases the protein level of NEDL2 but has no effect on the NEDL2 mRNA level. NEDL2 associates with mitotic spindles, and its protein level reaches a maximum in mitosis. The function of NEDL2 during mitosis is essential because NEDL2 depletion prolongs metaphase, and overexpression of NEDL2 induces chromosomal lagging. Elevated expression of NEDL2 protein and mRNA are both found in colon cancer and cervix cancer. We conclude that NEDL2 is a novel substrate of APC/C-Cdh1 as cells exit mitosis and functions as a regulator of the metaphase to anaphase transition. Its overexpression may contribute to tumorigenesis.
Collapse
Affiliation(s)
- Li Lu
- From the State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Regulation of APC(Cdh1) E3 ligase activity by the Fbw7/cyclin E signaling axis contributes to the tumor suppressor function of Fbw7. Cell Res 2013; 23:947-61. [PMID: 23670162 DOI: 10.1038/cr.2013.67] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/11/2013] [Accepted: 03/19/2013] [Indexed: 12/21/2022] Open
Abstract
Fbw7 and Cdh1 are substrate-recognition subunits of the SCF- and APC-type E3 ubiquitin ligases, respectively. There is emerging evidence suggesting that both Fbw7 and Cdh1 function as tumor suppressors by targeting oncoproteins for destruction. Loss of Fbw7, but not Cdh1, is frequently observed in various human tumors. However, it remains largely unknown how Fbw7 mechanistically functions as a tumor suppressor and whether there is a signaling crosstalk between Fbw7 and Cdh1. Here, we report that Fbw7-deficient cells not only display elevated expression levels of SCF(Fbw7) substrates, including cyclin E, but also have increased expression of various APC(Cdh1) substrates. We further defined cyclin E as the critical signaling link by which Fbw7 governs APC(Cdh1) activity, as depletion of cyclin E in Fbw7-deficient cells results in decreased expression of APC(Cdh1) substrates to levels comparable to those in wild-type (WT) cells. Conversely, ectopic expression of cyclin E recapitulates the aberrant APC(Cdh1) substrate expression observed in Fbw7-deficient cells. More importantly, 4A-Cdh1 that is resistant to Cdk2/cyclin E-mediated phosphorylation, but not WT-Cdh1, reversed the elevated expression of various APC(Cdh1) substrates in Fbw7-deficient cells. Overexpression of 4A-Cdh1 also resulted in retarded cell growth and decreased anchorage-independent colony formation. Altogether, we have identified a novel regulatory mechanism by which Fbw7 governs Cdh1 activity in a cyclin E-dependent manner. As a result, loss of Fbw7 can lead to aberrant increase in the expression of both SCF(Fbw7) and APC(Cdh1) substrates. Our study provides a better understanding of the tumor suppressor function of Fbw7, and suggests that Cdk2/cyclin E inhibitors could serve as effective therapeutic agents for treating Fbw7-deficient tumors.
Collapse
|
34
|
Cives M, Ciavarella S, Rizzo FM, De Matteo M, Dammacco F, Silvestris F. Bendamustine overcomes resistance to melphalan in myeloma cell lines by inducing cell death through mitotic catastrophe. Cell Signal 2013; 25:1108-17. [PMID: 23380051 DOI: 10.1016/j.cellsig.2013.01.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 01/25/2013] [Indexed: 12/01/2022]
Abstract
Melphalan has been a mainstay of multiple myeloma (MM) therapy for many years. However, following treatment with this alkylator, malignant plasma cells usually escape both apoptosis and cell cycle control, and acquire drug-resistance resulting in tumor progression. Bendamustine is being used in MM patients refractory to conventional DNA-damaging agents, although the mechanisms driving this lack of cross-resistance are still undefined. Here, we investigated the molecular pathway of bendamustine-induced cell death in melphalan-sensitive and melphalan-resistant MM cell lines. Bendamustine affected cell survival resulting in secondary necrosis, and prompted cell death primarily through caspase-2 activation. Also, bendamustine blocked the cell cycle in the G2/M phase and induced micronucleation, erratic chromosome spreading and mitotic spindle perturbations in melphalan-resistant MM cells. In these cells, both Aurora kinase A (AURKA) and Polo-like kinase-1 (PLK-1), key components of the spindle-assembly checkpoint, were down-regulated following incubation with bendamustine, whereas levels of Cyclin B1 increased as a consequence of the prolonged mitotic arrest induced by the drug. These findings indicate that, at least in vitro, bendamustine drives cell death by promoting mitotic catastrophe in melphalan-resistant MM cells. Hence, activation of this alternative pathway of cell death may be a novel approach to the treatment of apoptosis-resistant myelomas.
Collapse
Affiliation(s)
- Mauro Cives
- Department of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | | | | | | | | | | |
Collapse
|
35
|
Khattar V, Thottassery JV. Cks1: Structure, Emerging Roles and Implications in Multiple Cancers. ACTA ACUST UNITED AC 2013; 4:1341-1354. [PMID: 24563807 PMCID: PMC3930463 DOI: 10.4236/jct.2013.48159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Deregulation of the cell cycle results in loss of normal control mechanisms that prevent aberrant cell proliferation and cancer progression. Regulation of the cell cycle is a highly complex process with many layers of control. One of these mechanisms involves timely degradation of CDK inhibitors (CKIs) like p27Kip1 by the ubiquitin proteasomal system (UPS). Cks1 is a 9 kDa protein which is frequently overexpressed in different tumor subtypes, and has pleiotropic roles in cell cycle progression, many of which remain to be fully characterized. One well characterized molecular role of Cks1 is that of an essential adaptor that regulates p27Kip1 abundance by facilitating its interaction with the SCF-Skp2 E3 ligase which appends ubiquitin to p27Kip1 and targets it for degradation through the UPS. In addition, emerging research has uncovered p27Kip1-independent roles of Cks1 which have provided crucial insights into how it may be involved in cancer progression. We review here the structural features of Cks1 and their functional implications, and also some recently identified Cks1 roles and their involvement in breast and other cancers.
Collapse
Affiliation(s)
| | - Jaideep V Thottassery
- Southern Research Institute, Birmingham, USA ; University of Alabama Comprehensive Cancer Center, Birmingham, USA
| |
Collapse
|
36
|
Kwan PS, Lau CC, Chiu YT, Man C, Liu J, Tang KD, Wong YC, Ling MT. Daxx regulates mitotic progression and prostate cancer predisposition. Carcinogenesis 2012; 34:750-9. [PMID: 23239745 DOI: 10.1093/carcin/bgs391] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mitotic progression of mammalian cells is tightly regulated by the E3 ubiquitin ligase anaphase promoting complex (APC)/C. Deregulation of APC/C is frequently observed in cancer cells and is suggested to contribute to chromosome instability and cancer predisposition. In this study, we identified Daxx as a novel APC/C inhibitor frequently overexpressed in prostate cancer. Daxx interacts with the APC/C coactivators Cdc20 and Cdh1 in vivo, with the binding of Cdc20 dependent on the consensus destruction boxes near the N-terminal of the Daxx protein. Ectopic expression of Daxx, but not the D-box deleted mutant (DaxxΔD-box), inhibited the degradation of APC/Cdc20 and APC/Cdh1 substrates, leading to a transient delay in mitotic progression. Daxx is frequently upregulated in prostate cancer tissues; the expression level positively correlated with the Gleason score and disease metastasis (P = 0.027 and 0.032, respectively). Furthermore, ectopic expression of Daxx in a non-malignant prostate epithelial cell line induced polyploidy under mitotic stress. Our data suggest that Daxx may function as a novel APC/C inhibitor, which promotes chromosome instability during prostate cancer development.
Collapse
Affiliation(s)
- Pak Shing Kwan
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Haaß W, Stehle M, Nittka S, Giehl M, Schrotz-King P, Fabarius A, Hofmann WK, Seifarth W. The proteolytic activity of separase in BCR-ABL-positive cells is increased by imatinib. PLoS One 2012; 7:e42863. [PMID: 22870341 PMCID: PMC3411713 DOI: 10.1371/journal.pone.0042863] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/13/2012] [Indexed: 01/10/2023] Open
Abstract
Separase, an endopeptidase required for the separation of sister-chromatides in mitotic anaphase, triggers centriole disengagement during centrosome duplication. In cancer, separase is frequently overexpressed, pointing to a functional role as an aneuploidy promoter associated with centrosomal amplification and genomic instability. Recently, we have shown that centrosomal amplification and subsequent chromosomal aberrations are a hallmark of chronic myeloid leukemia (CML), increasing from chronic phase (CP) toward blast crisis (BC). Moreover, a functional linkage of p210BCR-ABL tyrosine kinase activity with centrosomal amplification and clonal evolution has been established in long-term cell culture experiments. Unexpectedly, therapeutic doses of imatinib (IM) did not counteract; instead induced similar centrosomal alterations in vitro. We investigated the influence of IM and p210BCR-ABL on Separase as a potential driver of centrosomal amplification in CML. Short-term cell cultures of p210BCR-ABL-negative (NHDF, UROtsa, HL-60, U937), positive (K562, LAMA-84) and inducible (U937p210BCR-ABL/c6 (Tet-ON)) human cell lines were treated with therapeutic doses of IM and analyzed by qRT-PCR, Western blot analysis and quantitative Separase activity assays. Decreased Separase protein levels were observed in all cells treated with IM in a dose dependent manner. Accordingly, in all p210BCR-ABL-negative cell lines, decreased proteolytic activity of Separase was found. In contrast, p210BCR-ABL-positive cells showed increased Separase proteolytic activity. This activation of Separase was consistent with changes in the expression levels of Separase regulators (Separase phosphorylation at serine residue 1126, Securin, CyclinB1 and PP2A). Our data suggest that regulation of Separase in IM-treated BCR-ABL-positive cells occurs on both the protein expression and the proteolytic activity levels. Activation of Separase proteolytic activity exclusively in p210BCR-ABL-positive cells during IM treatment may act as a driving force for centrosomal amplification, contributing to genomic instability, clonal evolution and resistance in CML.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Benzamides
- Blast Crisis/drug therapy
- Blast Crisis/enzymology
- Blast Crisis/genetics
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cyclin B1/genetics
- Cyclin B1/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Endopeptidases/genetics
- Endopeptidases/metabolism
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Genomic Instability/drug effects
- Genomic Instability/genetics
- HL-60 Cells
- Humans
- Imatinib Mesylate
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Phosphorylation/drug effects
- Phosphorylation/genetics
- Piperazines/pharmacology
- Protein Phosphatase 2/genetics
- Protein Phosphatase 2/metabolism
- Proteolysis
- Pyrimidines/pharmacology
- Securin
- Separase
- U937 Cells
Collapse
Affiliation(s)
- Wiltrud Haaß
- Department of Hematology and Oncology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Michael Stehle
- Department of Hematology and Oncology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Stefanie Nittka
- Department of Clinical Chemistry, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Michelle Giehl
- Department of Hematology and Oncology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Petra Schrotz-King
- National Center for Tumor Diseases (NCT), German Cancer Center (DKFZ), Heidelberg, Germany
| | - Alice Fabarius
- Department of Hematology and Oncology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Wolfgang Seifarth
- Department of Hematology and Oncology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
- * E-mail:
| |
Collapse
|
38
|
Wang LY, Kung HJ. Male germ cell-associated kinase is overexpressed in prostate cancer cells and causes mitotic defects via deregulation of APC/CCDH1. Oncogene 2012; 31:2907-18. [PMID: 21986944 PMCID: PMC3566783 DOI: 10.1038/onc.2011.464] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 08/02/2011] [Accepted: 09/06/2011] [Indexed: 11/09/2022]
Abstract
Male germ cell-associated kinase (MAK), a direct transcriptional target of androgen receptor (AR), is a co-activator of AR. In this study, we determined the activating mechanism of MAK and identified a previously unknown AR-independent role of MAK in mitosis. We found that MAK kinase activity requires dual phosphorylation of the conserved TDY motif and that the phosphorylation is dynamic during cell cycle. MAK associates with CDH1 (FZR1, fizzy/cell division cycle 20 related 1) and phosphorylates CDH1 at sites phosphorylated by cyclin-dependent kinases. When MAK is overexpressed, the binding of CDH1 to anaphase promoting complex/cyclosome decreased, resulting in an attenuation of anaphase-promoting complex/C ubiquitin ligase activity and the consequential stabilization of the CDH1 targets such as Aurora kinase A and Polo-like kinase 1. As such, overexpression of MAK leads to mitotic defects such as centrosome amplification and lagging chromosomes. Our immunohistochemistry result showed that MAK is overexpressed in prostate tumor tissues, suggesting a role of MAK in prostate carcinogenesis. Taken with our previous results, our data implicate MAK in both AR activation and chromosomal instability, acting in both early and late prostate cancer development.
Collapse
Affiliation(s)
- L-Y Wang
- Department of Biochemistry and Molecular Medicine and University of California Davis Cancer Center, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | | |
Collapse
|
39
|
Role of translationally controlled tumor protein in cancer progression. Biochem Res Int 2012; 2012:369384. [PMID: 22570787 PMCID: PMC3337558 DOI: 10.1155/2012/369384] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 02/17/2012] [Indexed: 12/14/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved and ubiquitously expressed protein in all eukaryotes-highlighting its important functions in the cell. Previous studies revealed that TCTP is implicated in many biological processes, including cell growth, tumor reversion, and induction of pluripotent stem cell. A recent study on the solution structure from fission yeast orthologue classifies TCTP under a family of small chaperone proteins. There is growing evidence in the literature that TCTP is a multifunctional protein and exerts its biological activity at the extracellular and intracellular levels. Although TCTP is not a tumor-specific protein, our research group, among several others, focused on the role(s) of TCTP in cancer progression. In this paper, we will summarize the current scientific knowledge of TCTP in different aspects, and the precise oncogenic mechanisms of TCTP will be discussed in detail.
Collapse
|
40
|
Kato T, Daigo Y, Aragaki M, Ishikawa K, Sato M, Kaji M. Overexpression of CDC20 predicts poor prognosis in primary non-small cell lung cancer patients. J Surg Oncol 2012; 106:423-30. [DOI: 10.1002/jso.23109] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/06/2012] [Indexed: 01/29/2023]
|
41
|
NAHA, a novel hydroxamic acid-derivative, inhibits growth and angiogenesis of breast cancer in vitro and in vivo. PLoS One 2012; 7:e34283. [PMID: 22479587 PMCID: PMC3315582 DOI: 10.1371/journal.pone.0034283] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 02/27/2012] [Indexed: 11/29/2022] Open
Abstract
Background We have recently synthesized novel N-alkylated amino acid-derived hydroxamate, 2-[Benzyl-(2-nitro-benzenesulfonyl)-amino]-N-hydroxy-3-methyl-N-propyl-butyramide (NAHA). Here, we evaluate the anticancer activity of NAHA against highly invasive human breast cancer cells MDA-MB-231 in vitro and in vivo. Methodology/Principal Findings Cell growth was evaluated by MTT and soft agar assays. Protein expression was determined by DNA microarray and Western blot analysis. Metastatic potential was evaluated by cell adhesion, migration, invasion, capillary morphogenesis, and ELISA assays. The anticancer activity in vivo was evaluated in mouse xenograft model. NAHA inhibited proliferation and colony formation of MDA-MB-231 cells together with the down-regulation of expression of Cdk2 and CDC20 proteins. NAHA inhibited cell adhesion, migration, and invasion through the suppression of secretion of uPA. NAHA suppressed secretion of VEGF from MDA-MB-231 cells and inhibited capillary morphogenesis of human aortic endothelial cells (HAECs). Finally, NAHA at 50 mg/kg was not toxic and decreased tumor volume and tumor weight in vivo. This suppression of tumor growth was associated with the inhibition of mitotic figures and induction of apoptosis, and the reduction of CD31 and VEGF positive cells in tumors. Conclusion NAHA could be a novel promising compound for the development of new drugs for the therapy of invasive breast cancers.
Collapse
|
42
|
Luo J, Liu X. Polo-like kinase 1, on the rise from cell cycle regulation to prostate cancer development. Protein Cell 2012; 3:182-97. [PMID: 22447658 PMCID: PMC4875424 DOI: 10.1007/s13238-012-2020-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Accepted: 02/04/2012] [Indexed: 01/19/2023] Open
Abstract
Polo-like kinase 1 (Plk1), a well-characterized member of serine/threonine kinases Plk family, has been shown to play pivotal roles in mitosis and cytokinesis in eukaryotic cells. Recent studies suggest that Plk1 not only controls the process of mitosis and cytokinesis, but also, going beyond those previously described functions, plays critical roles in DNA replication and Pten null prostate cancer initiation. In this review, we briefly summarize the functions of Plk1 in mitosis and cytokinesis, and then mainly focus on newly discovered functions of Plk1 in DNA replication and in Pten-null prostate cancer initiation. Furthermore, we briefly introduce the architectures of human and mouse prostate glands and the possible roles of Plk1 in human prostate cancer development. And finally, the newly chemotherapeutic development of small-molecule Plk1 inhibitors to target Plk1 in cancer treatment and their translational studies are also briefly reviewed.
Collapse
Affiliation(s)
- Jijing Luo
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907 USA
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907 USA
| |
Collapse
|
43
|
Penas C, Ramachandran V, Ayad NG. The APC/C Ubiquitin Ligase: From Cell Biology to Tumorigenesis. Front Oncol 2012; 1:60. [PMID: 22655255 PMCID: PMC3356048 DOI: 10.3389/fonc.2011.00060] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/22/2011] [Indexed: 01/15/2023] Open
Abstract
The ubiquitin proteasome system (UPS) is required for normal cell proliferation, vertebrate development, and cancer cell transformation. The UPS consists of multiple proteins that work in concert to target a protein for degradation via the 26S proteasome. Chains of an 8.5-kDa protein called ubiquitin are attached to substrates, thus allowing recognition by the 26S proteasome. Enzymes called ubiquitin ligases or E3s mediate specific attachment to substrates. Although there are over 600 different ubiquitin ligases, the Skp1-Cullin-F-box (SCF) complexes and the anaphase promoting complex/cyclosome (APC/C) are the most studied. SCF involvement in cancer has been known for some time while APC/C's cancer role has recently emerged. In this review we will discuss the importance of APC/C to normal cell proliferation and development, underscoring its possible contribution to transformation. We will also examine the hypothesis that modulating a specific interaction of the APC/C may be therapeutically attractive in specific cancer subtypes. Finally, given that the APC/C pathway is relatively new as a cancer target, therapeutic interventions affecting APC/C activity may be beneficial in cancers that are resistant to classical chemotherapy.
Collapse
Affiliation(s)
- Clara Penas
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine Miami, FL, USA
| | | | | |
Collapse
|
44
|
Qiao D, Meyer K, Friedl A. Glypican-1 stimulates Skp2 autoinduction loop and G1/S transition in endothelial cells. J Biol Chem 2011; 287:5898-909. [PMID: 22203671 DOI: 10.1074/jbc.m111.325282] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The heparan sulfate proteoglycan glypican-1 (GPC1) is involved in tumorigenesis and angiogenesis and is overexpressed frequently in tumor and endothelial cells (ECs) in human gliomas. We demonstrated previously that in brain EC, GPC1 regulates mitotic cyclins and securin as well as mitosis and that GPC1 is required for progression through the cell cycle. To characterize the molecular mechanism underlying cell cycle regulation by GPC1, we systematically investigated its effects on key G(1)/S checkpoint regulators and on major signaling pathways reportedly activated by Dally (Division abnormally delayed) the Drosophila GPC1 homologue. We found that elevated GPC1 affected a wide range of G(1)/S checkpoint regulators, leading to inactivation of the G(1)/S checkpoint and increased S phase entry, apparently by activating the mitogen-independent Skp2 autoinduction loop. Specifically, GPC1 suppressed CDK inhibitors (CKIs), including p21, p27, p16, and p19, and the D cyclins, and induced CDK2 and Skp2. GPC1 may trigger the Skp2 autoinduction loop at least partially by suppressing p21 transcription as knockdown of p21 by RNAi can mimic the effect of GPC1 on the cell cycle regulators related to the loop. Moreover, multiple mitogenic signaling pathways, including ERK MAPK, Wnt and BMP signaling, were significantly stimulated by GPC1 as has been reported for Dally in Drosophila. Notably, the c-Myc oncoprotein, which is frequently up-regulated by both ERK and Wnt signaling and functions as a potent transcription repressor for CKIs as well as D cyclins, was also significantly induced by GPC1. These findings provide mechanistic insights into how GPC1 regulates the cell cycle and proliferation.
Collapse
Affiliation(s)
- Dianhua Qiao
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | | | | |
Collapse
|
45
|
Jiang J, Jedinak A, Sliva D. Ganodermanontriol (GDNT) exerts its effect on growth and invasiveness of breast cancer cells through the down-regulation of CDC20 and uPA. Biochem Biophys Res Commun 2011; 415:325-9. [PMID: 22033405 DOI: 10.1016/j.bbrc.2011.10.055] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 10/11/2011] [Indexed: 01/11/2023]
Abstract
Ganoderma lucidum is a medicinal mushroom that has been recognized by Traditional Chinese Medicine (TCM). Although some of the direct anticancer activities are attributed to the presence of triterpenes-ganoderic and lucidenic acids-the activity of other compounds remains elusive. Here we show that ganodermanontriol (GDNT), a Ganoderma alcohol, specifically suppressed proliferation (anchorage-dependent growth) and colony formation (anchorage-independent growth) of highly invasive human breast cancer cells MDA-MB-231. GDNT suppressed expression of the cell cycle regulatory protein CDC20, which is over-expressed in precancerous and breast cancer cells compared to normal mammary epithelial cells. Moreover, we found that CDC20 is over-expressed in tumors when compared to the tissue surrounding the tumor in specimens from breast cancer patients. GDNT also inhibited invasive behavior (cell adhesion, cell migration, and cell invasion) through the suppression of secretion of urokinase-plasminogen activator (uPA) and inhibited expression of uPA receptor. In conclusion, mushroom GDNT is a natural agent that has potential as a therapy for invasive breast cancers.
Collapse
Affiliation(s)
- Jiahua Jiang
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN, USA
| | | | | |
Collapse
|
46
|
Puliyappadamba VT, Wu W, Bevis D, Zhang L, Polin L, Kilkuskie R, Finley RL, Larsen SD, Levi E, Miller FR, Wali A, Rishi AK. Antagonists of anaphase-promoting complex (APC)-2-cell cycle and apoptosis regulatory protein (CARP)-1 interaction are novel regulators of cell growth and apoptosis. J Biol Chem 2011; 286:38000-38017. [PMID: 21903591 DOI: 10.1074/jbc.m111.222398] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CARP-1/CCAR1, a perinuclear phosphoprotein, is a regulator of cell growth and apoptosis signaling. Although CARP-1 is a regulator of chemotherapy-dependent apoptosis, it is also a part of the NF-κB proteome and a co-activator of steroid/thyroid nuclear receptors as well as β-catenin signaling. Our yeast two-hybrid screen revealed CARP-1 binding with the anaphase-promoting complex/cyclosome E3 ubiquitin ligase component APC-2 protein. CARP-1 also binds with anaphase-promoting complex/cyclosome co-activators Cdc20 and Cdh1. Following mapping of the minimal epitopes involved in CARP-1 binding with APC-2, a fluorescence polarization assay was established that indicated a dissociation constant (K(d)) of 480 nm for CARP-1/APC-2 binding. Fluorescence polarization assay-based high throughput screening of a chemical library yielded several small molecule antagonists of CARP-1/APC-2 binding, termed CARP-1 functional mimetics. CFM-4 (1(2-chlorobenzyl)-5'-phenyl-3'H-spiro[indoline-3,2'-[1,3,4]thiadiazol]-2-one), a lead compound, binds with and stimulates CARP-1 expression. CFM-4 prevents CARP-1 binding with APC-2, causes G(2)M cell cycle arrest, and induces apoptosis with an IC(50) range of 10-15 μm. Apoptosis signaling by CFM-4 involves activation of caspase-8 and -9 and caspase-mediated ubiquitin-proteasome pathway-independent loss of cyclin B1 and Cdc20 proteins. Depletion of CARP-1, however, interferes with CFM-4-dependent cell growth inhibition, activation of caspases, and apoptosis. Because CFM-4 also suppresses growth of drug-resistant human breast cancer cells without affecting the growth of human breast epithelial MCF-10A cells, elevating CARP-1 by CFM-4 and consequent apoptosis could in principle be exploited to further elucidate, and perhaps effectively target, often deregulated cell cycle pathways in pathological conditions, including cancer.
Collapse
Affiliation(s)
| | - Wenjuan Wu
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Debra Bevis
- Michigan High-throughput Screening Center, Kalamazoo Valley Community College, Kalamazoo, Michigan 49003
| | - Liyue Zhang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; John D. Dingell Veterans Affairs Medical Center, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Lisa Polin
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Robert Kilkuskie
- Michigan High-throughput Screening Center, Kalamazoo Valley Community College, Kalamazoo, Michigan 49003
| | - Russell L Finley
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Scott D Larsen
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Edi Levi
- John D. Dingell Veterans Affairs Medical Center, Wayne State University School of Medicine, Detroit, Michigan 48201; Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Fred R Miller
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201; Breast Cancer Program, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Anil Wali
- John D. Dingell Veterans Affairs Medical Center, Wayne State University School of Medicine, Detroit, Michigan 48201; Department of Surgery, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Arun K Rishi
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; John D. Dingell Veterans Affairs Medical Center, Wayne State University School of Medicine, Detroit, Michigan 48201; Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan 48201; Breast Cancer Program, Wayne State University School of Medicine, Detroit, Michigan 48201.
| |
Collapse
|
47
|
Lipkowitz S, Weissman AM. RINGs of good and evil: RING finger ubiquitin ligases at the crossroads of tumour suppression and oncogenesis. Nat Rev Cancer 2011; 11:629-43. [PMID: 21863050 PMCID: PMC3542975 DOI: 10.1038/nrc3120] [Citation(s) in RCA: 306] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ubiquitin-proteasome system has numerous crucial roles in physiology and pathophysiology. Fundamental to the specificity of this system are ubiquitin-protein ligases (E3s). Of these, the majority are RING finger and RING finger-related E3s. Many RING finger E3s have roles in processes that are central to the maintenance of genomic integrity and cellular homeostasis, such as the anaphase promoting complex/cyclosome (APC/C), the SKP1-cullin 1-F-box protein (SCF) E3s, MDM2, BRCA1, Fanconi anaemia proteins, CBL proteins, von Hippel-Lindau tumour suppressor (VHL) and SIAH proteins. As a result, many RING finger E3s are implicated in either the suppression or the progression of cancer. This Review summarizes current knowledge in this area.
Collapse
Affiliation(s)
- Stanley Lipkowitz
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
48
|
Huang X, Summers MK, Pham V, Lill JR, Liu J, Lee G, Kirkpatrick DS, Jackson PK, Fang G, Dixit VM. Deubiquitinase USP37 is activated by CDK2 to antagonize APC(CDH1) and promote S phase entry. Mol Cell 2011; 42:511-23. [PMID: 21596315 DOI: 10.1016/j.molcel.2011.03.027] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 03/07/2011] [Accepted: 03/30/2011] [Indexed: 11/26/2022]
Abstract
Cell cycle progression requires the E3 ubiquitin ligase anaphase-promoting complex (APC/C), which uses the substrate adaptors CDC20 and CDH1 to target proteins for proteasomal degradation. The APC(CDH1) substrate cyclin A is critical for the G1/S transition and, paradoxically, accumulates even when APC(CDH1) is active. We show that the deubiquitinase USP37 binds CDH1 and removes degradative polyubiquitin from cyclin A. USP37 was induced by E2F transcription factors in G1, peaked at G1/S, and was degraded in late mitosis. Phosphorylation of USP37 by CDK2 stimulated its full activity. USP37 overexpression caused premature cyclin A accumulation in G1 and accelerated S phase entry, whereas USP37 knockdown delayed these events. USP37 was inactive in mitosis because it was no longer phosphorylated by CDK2. Indeed, it switched from an antagonist to a substrate of APC(CDH1) and was modified with degradative K11-linked polyubiquitin.
Collapse
Affiliation(s)
- Xiaodong Huang
- Department of Physiological Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mora-Santos M, Limón-Mortés MC, Giráldez S, Herrero-Ruiz J, Sáez C, Japón MÁ, Tortolero M, Romero F. Glycogen synthase kinase-3beta (GSK3beta) negatively regulates PTTG1/human securin protein stability, and GSK3beta inactivation correlates with securin accumulation in breast tumors. J Biol Chem 2011; 286:30047-56. [PMID: 21757741 DOI: 10.1074/jbc.m111.232330] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PTTG1, also known as securin, is an inactivating partner of separase, the major effector for chromosome segregation during mitosis. At the metaphase-to-anaphase transition, securin is targeted for proteasomal destruction by the anaphase-promoting complex or cyclosome, allowing activation of separase. In addition, securin is overexpressed in metastatic or genomically instable tumors, suggesting a relevant role for securin in tumor progression. Stability of securin is regulated by phosphorylation; some phosphorylated forms are degraded out of mitosis, by the action of the SKP1-CUL1-F-box protein (SCF) complex. The kinases targeting securin for proteolysis have not been identified, and mechanistic insight into the cause of securin accumulation in human cancers is lacking. Here, we demonstrate that glycogen synthase kinase-3β (GSK3β) phosphorylates securin to promote its proteolysis via SCF(βTrCP) E3 ubiquitin ligase. Importantly, a strong correlation between securin accumulation and GSK3β inactivation was observed in breast cancer tissues, indicating that GSK3β inactivation may account for securin accumulation in breast cancers.
Collapse
Affiliation(s)
- Mar Mora-Santos
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Eguren M, Manchado E, Malumbres M. Non-mitotic functions of the Anaphase-Promoting Complex. Semin Cell Dev Biol 2011; 22:572-8. [PMID: 21439391 DOI: 10.1016/j.semcdb.2011.03.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Accepted: 03/16/2011] [Indexed: 02/03/2023]
Abstract
The Anaphase-Promoting Complex or Cyclosome (APC/C) is an E3 ubiquitin ligase whose activation requires the binding of a cofactor, either Cdc20 or Cdh1. While APC/C-Cdc20 is a major player during mitotic exit, APC/C-Cdh1 plays a central role in maintaining quiescence and controlling the onset of DNA replication. In addition, APC/C-Cdh1 is essential for endoreduplication, a process in which several rounds of DNA synthesis occur without mitosis. Recent data suggest that the APC/C is also involved in differentiation and metabolism, and plays important roles in postmitotic cells such as neurons. Thus, the APC/C is not only critical for anaphase onset but also regulates many other cellular processes during G1/S or in quiescent cells.
Collapse
Affiliation(s)
- Manuel Eguren
- Cell Division and Cancer Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | | |
Collapse
|