1
|
Chen YJC, Bhaskara GB, Lu Y, Lin K, Dent SYR. The SAGA acetyltransferase module is required for the maintenance of MAF and MYC oncogenic gene expression programs in multiple myeloma. Genes Dev 2024; 38:738-754. [PMID: 39168636 PMCID: PMC11444170 DOI: 10.1101/gad.351789.124] [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: 04/08/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024]
Abstract
Despite recent advances in therapeutic treatments, multiple myeloma (MM) remains an incurable malignancy. Epigenetic factors contribute to the initiation, progression, relapse, and clonal heterogeneity in MM, but our knowledge on epigenetic mechanisms underlying MM development is far from complete. The SAGA complex serves as a coactivator in transcription and catalyzes acetylation and deubiquitylation. Analyses of data sets in the Cancer Dependency Map Project revealed that many SAGA components are selective dependencies in MM. To define SAGA-specific functions, we focused on ADA2B, the only subunit in the lysine acetyltransferase (KAT) module that specifically functions in SAGA. Integration of RNA sequencing (RNA-seq), assay for transposase-accessible chromatin with sequencing (ATAC-seq), and cleavage under targets and release using nuclease assay (CUT&RUN) results identified pathways directly regulated by ADA2B including MTORC1 signaling and oncogenic programs driven by MYC, E2F, and MM-specific MAF. We discovered that ADA2B is recruited to MAF and MYC gene targets, and that MAF shares a majority of its targets with MYC in MM cells. Furthermore, we found that the SANT domain of ADA2B is required for interaction with both GCN5 and PCAF acetyltransferases, incorporation into SAGA, and ADA2B protein stability. Our findings uncover previously unknown SAGA KAT module-dependent mechanisms controlling MM cell growth, revealing a vulnerability that might be exploited for future development of MM therapy.
Collapse
Affiliation(s)
- Ying-Jiun C Chen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA;
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Govinal Badiger Bhaskara
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Sharon Y R Dent
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA;
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| |
Collapse
|
2
|
Chen YJC, Bhaskara GB, Lu Y, Lin K, Dent SYR. The SAGA acetyltransferase module is required for the maintenance of MAF and MYC oncogenic gene expression programs in multiple myeloma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586811. [PMID: 38585845 PMCID: PMC10996596 DOI: 10.1101/2024.03.26.586811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Despite recent advances in therapeutic treatments, multiple myeloma (MM) remains an incurable malignancy. Epigenetic factors contribute to the initiation, progression, relapse, and clonal heterogeneity in MM, but our knowledge on epigenetic mechanisms underlying MM development is far from complete. The SAGA complex serves as a coactivator in transcription and catalyzes acetylation and deubiquitylation. Analyses of datasets in the Cancer Dependency Map Project revealed many SAGA components are selective dependencies in MM. To define SAGA-specific functions, we focused on ADA2B, the only subunit in the lysine acetyltransferase (KAT) module that specifically functions in SAGA. Integration of RNA-seq, ATAC-seq, and CUT&RUN results identified pathways directly regulated by ADA2B include MTORC1 signaling, MYC, E2F, and MM-specific MAF oncogenic programs. We discovered that ADA2B is recruited to MAF and MYC gene targets, and that MAF shares a majority of its targets with MYC in MM cells. Furthermore, we found the SANT domain of ADA2B is required for interaction with both GCN5 and PCAF acetyltransferases, incorporation into SAGA, and ADA2B protein stability. Our findings uncover previously unknown SAGA KAT module-dependent mechanisms controlling MM cell growth, revealing a vulnerability that might be exploited for future development of MM therapy.
Collapse
Affiliation(s)
- Ying-Jiun C. Chen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Govinal Badiger Bhaskara
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sharon Y. R. Dent
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
3
|
Zhang Z, Li M, Lin P, Ren Y, He Y, Wang S, Xu Y, Cao B, Wang G, Moran MF, Mao X. The ubiquitin ligase HERC4 suppresses MafA transcriptional activity triggered by GSK3β in myeloma by atypical K63-linked polyubiquitination. J Biol Chem 2023; 299:104675. [PMID: 37028761 DOI: 10.1016/j.jbc.2023.104675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
MafA and c-Maf are close members of the Maf transcription factor family and indicators of poor prognosis of multiple myeloma (MM). Our previous study finds that the ubiquitin ligase HERC4 induces c-Maf degradation but stabilizes MafA, and the mechanism is elusive. In the present study we find that HERC4 interacts with MafA and mediates its K63-linked polyubiquitination at K33. Moreover, HERC4 inhibits MafA phosphorylation and its transcriptional activity triggered by glycogen synthase kinase 3β (GSK3β). The K33R MafA variant prevents HERC4 from inhibiting MafA phosphorylation and increases MafA transcriptional activity. Further analyses reveal that MafA can also activate the STAT3 signaling but it is suppressed by HERC4. Lastly, we demonstrate that lithium chloride, a GSK3β inhibitor, can upregulate HERC4 and synergizes dexamethasone, a typical anti-MM drug, in inhibiting MM cell proliferation and xenograft growth in nude mice. These findings thus highlight a novel regulation of MafA oncogenic activity in MM and provide the rationale by targeting HERC4/GSK3β/MafA for the treatment of MM.
Collapse
Affiliation(s)
- Zubin Zhang
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Mei Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215025, China
| | - Peng Lin
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ying Ren
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yuanming He
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Siyu Wang
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yujia Xu
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Biyin Cao
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Guanghui Wang
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Michael F Moran
- The Department of Molecular Genetics, The University of Toronto, Toronto, ON, M5G 0A4, Canada
| | - Xinliang Mao
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsychoDiseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| |
Collapse
|
4
|
He YM, Zhou XM, Jiang SY, Zhang ZB, Cao BY, Liu JB, Zeng YY, Zhao J, Mao XL. TRIM25 activates AKT/mTOR by inhibiting PTEN via K63-linked polyubiquitination in non-small cell lung cancer. Acta Pharmacol Sin 2022; 43:681-691. [PMID: 33931764 PMCID: PMC8888698 DOI: 10.1038/s41401-021-00662-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/17/2021] [Indexed: 12/21/2022]
Abstract
The PTEN/AKT/mTOR signaling pathway is frequently dysregulated in non-small cell lung cancer (NSCLC), but the mechanisms are not well-understood. The present study found that the ubiquitin ligase TRIM25 is highly expressed in NSCLC tissues and promotes NSCLC cell survival and tumor growth. Mechanistic studies revealed that TRIM25 binds to PTEN and mediates its K63-linked ubiquitination at K266. This modification prevents the plasma membrane translocation of PTEN and reduces its phosphatase activity therefore accumulating PI(3,4,5)P3. TRIM25 thus activates the AKT/mTOR signaling. Moreover, we found that the antibacterial nitroxoline can activate PTEN by reducing its K63-linked polyubiquitination and sensitizes NSCLC to cisplatin-induced apoptosis. This study thus identified a novel modulation on the PTEN signaling pathway by TRIM25 and provides a potential target for NSCLC treatment.
Collapse
Affiliation(s)
- Yuan-ming He
- grid.410737.60000 0000 8653 1072Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436 China ,grid.263761.70000 0001 0198 0694Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Xiu-min Zhou
- grid.429222.d0000 0004 1798 0228Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215106 China
| | - Shuo-yi Jiang
- grid.410737.60000 0000 8653 1072Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436 China ,grid.263761.70000 0001 0198 0694Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Zu-bin Zhang
- grid.263761.70000 0001 0198 0694Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Bi-yin Cao
- grid.263761.70000 0001 0198 0694Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Jin-bao Liu
- grid.410737.60000 0000 8653 1072Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| | - Yuan-ying Zeng
- grid.440227.70000 0004 1758 3572Department of Oncology, Suzhou Municipal Hospital, Suzhou, 215100 China
| | - Jun Zhao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China. .,Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215106, China.
| | - Xin-liang Mao
- grid.410737.60000 0000 8653 1072Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| |
Collapse
|
5
|
He Y, Jiang S, Mao C, Zheng H, Cao B, Zhang Z, Zhao J, Zeng Y, Mao X. The deubiquitinase USP10 restores PTEN activity and inhibits non-small cell lung cancer cell proliferation. J Biol Chem 2021; 297:101088. [PMID: 34416231 PMCID: PMC8429974 DOI: 10.1016/j.jbc.2021.101088] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 11/25/2022] Open
Abstract
The phosphatase and tensin homolog deleted on chromosome 10 (PTEN) protein is a key player in tumorigenesis of non–small cell lung cancer (NSCLC) and was recently found to be inactivated by tripartite motif containing 25 (TRIM25)–mediated K63-linked polyubiquitination. However, the deubiquitinase (Dub) coordinate TRIM25 in PTEN ubiquitination is still elusive. In the present study, we found that this K63-linked polyubiquitination could be ablated by the ubiquitin-specific protease 10 (USP10) in a screen against a panel of Dubs. We found using coimmununoprecipitation/immunoblotting that USP10 interacted with PTEN and reduced the K63-linked polyubiquitination of PTEN mediated by TRIM25 in NSCLC cells. Moreover, USP10, but not its inactive C424A deubiquitinating mutant or other Dubs, abolished PTEN from K63-linked polyubiquitination mediated by TRIM25. In contrast to TRIM25, USP10 restored PTEN phosphatase activity and reduced the production of the secondary messenger phosphatidylinositol-3,4,5-trisphosphate, thereby inhibiting AKT/mammalian target of rapamycin progrowth signaling transduction in NSCLC cells. Moreover, USP10 was downregulated in NSCLC cell lines and primary tissues, whereas TRIM25 was upregulated. Consistent with its molecular activity, re-expression of USP10 suppressed NSCLC cell proliferation and migration, whereas knockout of USP10 promoted NSCLC cell proliferation and migration. In conclusion, the present study demonstrates that USP10 coordinates TRIM25 to modulate PTEN activity. Specifically, USP10 activates PTEN by preventing its K63-linked polyubiquitination mediated by TRIM25 and suppresses the AKT/mammalian target of rapamycin signaling pathway, thereby inhibiting NSCLC proliferation, indicating that it may be a potential drug target for cancer treatment.
Collapse
Affiliation(s)
- Yuanming He
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital, Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Shuoyi Jiang
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital, Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Chenyu Mao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hui Zheng
- Institute of Biomedical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Biyin Cao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Zubin Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuanying Zeng
- Department of Oncology, Suzhou Municipal Hospital, Suzhou, China.
| | - Xinliang Mao
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital, Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
6
|
Jiang S, Wang X, He Y, Huang H, Cao B, Zhang Z, Liu J, Wang Q, Huang Z, Mao X. Suppression of USP7 induces BCR-ABL degradation and chronic myelogenous leukemia cell apoptosis. Cell Death Dis 2021; 12:456. [PMID: 33963175 PMCID: PMC8105359 DOI: 10.1038/s41419-021-03732-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/25/2022]
Abstract
Chronic myelogenous leukemia (CML) is a clonal malignancy of hematopoietic stem cells featured with the fusion protein kinase BCR-ABL. To elicit the mechanism underlying BCR-ABL stability, we perform a screen against a panel of deubiquitinating enzymes (DUBs) and find that the ubiquitin-specific protease 7 (USP7) drastically stabilizes the BCR-ABL fusion protein. Further studies show that USP7 interacts with BCR-ABL and blocks its polyubiquitination and degradation. Moreover, USP7 knockdown triggers BCR-ABL degradation and suppresses its downstream signaling transduction. In line with this finding, genetic or chemical inhibition of USP7 leads to BCR-ABL protein degradation, suppresses BCR/ABL signaling, and induces CML cell apoptosis. Furthermore, we find the antimalarial artesunate (ART) significantly inhibits USP7/BCR-ABL interaction, thereby promoting BCR-ABL degradation and inducing CML cell death. This study thus identifies USP7 as a putative Dub of BCR-ABL and provides a rationale in targeting USP7/BCR-ABL for the treatment of CML.
Collapse
Affiliation(s)
- Shuoyi Jiang
- Department of Hematology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.,Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China.,Department of Pharmacology, Soochow University, Jiangsu, 215123, P. R. China
| | - Xiaoge Wang
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yuanming He
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China.,Department of Pharmacology, Soochow University, Jiangsu, 215123, P. R. China
| | - Hongbiao Huang
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Biyin Cao
- Department of Pharmacology, Soochow University, Jiangsu, 215123, P. R. China
| | - Zubin Zhang
- Department of Pharmacology, Soochow University, Jiangsu, 215123, P. R. China
| | - Jinbao Liu
- Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Qi Wang
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhenqian Huang
- Department of Hematology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Xinliang Mao
- Department of Hematology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China. .,Guangdong and Guangzhou Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China. .,Department of Pharmacology, Soochow University, Jiangsu, 215123, P. R. China. .,Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
7
|
Xu Y, Xu M, Tong J, Tang X, Chen J, Chen X, Zhang Z, Cao B, Stewart AK, Moran MF, Wu D, Mao X. Targeting the Otub1/c-Maf axis for the treatment of multiple myeloma. Blood 2021; 137:1478-1490. [PMID: 32842143 DOI: 10.1182/blood.2020005199] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
The oncogenic transcription factor c-Maf has been proposed as an ideal therapeutic target for multiple myeloma (MM), but how to achieve it is still elusive. In the present study, we found the Otub1/c-Maf axis could be a potential target. Otub1, an OTU family deubiquitinase, was found to interact with c-Maf by mass spectrometry. Otub1 abrogates c-Maf K48-linked polyubiquitination, thus preventing its degradation and enhancing its transcriptional activity. Specifically, this deubiquitinating activity depends on its Lys71 and the N terminus but is independent of UBE2O, a known E2 of c-Maf. Otub1 promotes MM cell survival and MM tumor growth. In contrast, silence of Otub1 leads to c-Maf degradation and c-Maf-expressing MM cell apoptosis. Therefore, the Otub1/c-Maf axis could be a therapeutic target of MM. In order to explore this concept, we performed a c-Maf recognition element-driven luciferase-based screen against US Food and Drug Administration-approved drugs and natural products, from which the generic cardiac glycoside lanatoside C (LanC) is found to prevent c-Maf deubiquitination and induces its degradation by disrupting the interaction of Otub1 and c-Maf. Consequently, LanC inhibits c-Maf transcriptional activity, induces c-Maf-expressing MM cell apoptosis, and suppresses MM growth and prolongs overall survival of model mice, but without apparent toxicity. Therefore, the present study identifies Otub1 as a novel deubiquitinase of c-Maf and establishes that the Otub1/c-Maf axis is a potential therapeutic target for MM.
Collapse
Affiliation(s)
- Yujia Xu
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital-Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, People's Republic of China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Min Xu
- Department of Hematology, Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Jiefei Tong
- Program in Cell Biology, The Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Xiaowen Tang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Suzhou, China
| | - Jinhao Chen
- Department of Hematology, Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Xuehan Chen
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Zubin Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Biyin Cao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | | | - Michael F Moran
- Program in Cell Biology, The Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Suzhou, China
| | - Xinliang Mao
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital-Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, People's Republic of China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, People's Republic of China
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Sun T, Xu Y, Xu Z, Cao B, Zhang Z, Wang Q, Kong Y, Mao X. Inhibition of the Otub1/c-Maf axis by the herbal acevaltrate induces myeloma cell apoptosis. Cell Commun Signal 2021; 19:24. [PMID: 33627137 PMCID: PMC7905600 DOI: 10.1186/s12964-020-00676-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The oncogenic transcript factor c-Maf is stabilized by the deubiquitinase Otub1 and promotes myeloma cell proliferation and confers to chemoresistance. Inhibition of the Otub1/c-Maf axis is a promising therapeutic target, but there are no inhibitors reported on this specific axis. METHODS A luciferase assay was applied to screen potential inhibitors of Otub1/c-Maf. Annexin V staining/flow cytometry was applied to evaluate cell apoptosis. Immunoprecipitation was applied to examine protein ubiquitination and interaction. Xenograft models in nude mice were used to evaluate anti-myeloma activity of AVT. RESULTS Acevaltrate (AVT), isolated from Valeriana glechomifolia, was identified based on a bioactive screen against the Otub1/c-Maf/luciferase system. AVT disrupts the interaction of Otub1/c-Maf thus inhibiting Otub1 activity and leading to c-Maf polyubiquitination and subsequent degradation in proteasomes. Consistently, AVT inhibits c-Maf transcriptional activity and downregulates the expression of its target genes key for myeloma growth and survival. Moreover, AVT displays potent anti-myeloma activity by triggering myeloma cell apoptosis in vitro and impairing myeloma xenograft growth in vivo but presents no marked toxicity. CONCLUSIONS The natural product AVT inhibits the Otub1/c-Maf axis and displays potent anti-myeloma activity. Given its great safety and efficacy, AVT could be further developed for MM treatment. Video Abstract.
Collapse
Affiliation(s)
- Tong Sun
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405 People’s Republic of China
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215100 Jiangsu People’s Republic of China
| | - Yujia Xu
- Department of Pharmacology, Soochow University, Suzhou, 215123 Jiangsu People’s Republic of China
| | - Zhuan Xu
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215100 Jiangsu People’s Republic of China
| | - Biyin Cao
- Department of Pharmacology, Soochow University, Suzhou, 215123 Jiangsu People’s Republic of China
| | - Zubin Zhang
- Department of Pharmacology, Soochow University, Suzhou, 215123 Jiangsu People’s Republic of China
| | - Qi Wang
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405 People’s Republic of China
| | - Yan Kong
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215100 Jiangsu People’s Republic of China
| | - Xinliang Mao
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405 People’s Republic of China
- Department of Pharmacology, Soochow University, Suzhou, 215123 Jiangsu People’s Republic of China
- Guangdong Institute of Cardiovascular Diseases & Guangdong Key Lab for Protein Modifications and Degradation, The Second Affiliated Hospital & School of Basic Medicine, Guangzhou Medical University , Guangzhou, 511436 People’s Republic of China
| |
Collapse
|
9
|
Xu Y, Sun T, Zeng K, Xu M, Chen J, Xu X, Zhang Z, Cao B, Tang X, Wu D, Kong Y, Zeng Y, Mao X. Anti-bacterial and anti-viral nanchangmycin displays anti-myeloma activity by targeting Otub1 and c-Maf. Cell Death Dis 2020; 11:818. [PMID: 32999280 PMCID: PMC7527563 DOI: 10.1038/s41419-020-03017-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022]
Abstract
As a deubiqutinase Otub1 stabilizes and promotes the oncogenic activity of the transcription factor c-Maf in multiple myeloma (MM), a malignancy of plasma cells. In the screen for bioactive inhibitors of the Otub1/c-Maf axis for MM treatment, nanchangmycin (Nam), a polyketide antibiotic, was identified to suppress c-Maf activity in the presence of Otub1. By suppressing Otub1, Nam induces c-Maf polyubiquitination and subsequent degradation in proteasomes but does not alter its mRNA level. Consistently, Nam downregulates the expression of CCND2, ARK5, and ITGB7, the downstream genes regulated by c-Maf, and promotes MM cell apoptosis as evidenced by PARP and Caspase-3 cleavage, as well as Annexin V staining. In line with the hypothesis, overexpression of Otub1 partly rescues Nam-induced MM cell apoptosis, and interestingly, when Otub1 is knocked down, Nam-decreased MM cell survival is also partly ablated, suggesting Otub1 is essential for Nam anti-MM activity. Nam also displays potent anti-MM activity synergistically with Doxorubicin or lenalidomide. In the in vivo assays, Nam almost completely suppresses the growth of MM xenografts in nude mice at low dosages but it shows no toxicity. Given its safety and efficacy, Nam has a potential for MM treatment by targeting the Otub1/c-Maf axis.
Collapse
Affiliation(s)
- Yujia Xu
- Guangdong Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital; Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Tong Sun
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215100, P. R. China
| | - Kun Zeng
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Min Xu
- Department of Hematology, Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215620, China
| | - Jinhao Chen
- Department of Hematology, Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215620, China
| | - Xiaofeng Xu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215100, P. R. China
| | - Zubin Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Biyin Cao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Xiaowen Tang
- Department of Urology, Nanjing Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210002, P. R. China
| | - Depei Wu
- Department of Urology, Nanjing Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, 210002, P. R. China
| | - Yan Kong
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215100, P. R. China
| | - Yuanying Zeng
- Department of Oncology, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215100, P. R. China.
| | - Xinliang Mao
- Guangdong Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital; Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China.
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
| |
Collapse
|
10
|
Hsu CY, Fu SH, Chien MW, Liu YW, Chen SJ, Sytwu HK. Post-Translational Modifications of Transcription Factors Harnessing the Etiology and Pathophysiology in Colonic Diseases. Int J Mol Sci 2020; 21:ijms21093207. [PMID: 32369982 PMCID: PMC7246881 DOI: 10.3390/ijms21093207] [Citation(s) in RCA: 10] [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: 03/31/2020] [Revised: 04/23/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023] Open
Abstract
Defects in mucosal immune balance can lead to colonic diseases such as inflammatory bowel diseases and colorectal cancer. With the advancement of understanding for the immunological and molecular basis of colonic disease, therapies targeting transcription factors have become a potential approach for the treatment of colonic disease. To date, the biomedical significance of unique post-translational modifications on transcription factors has been identified, including phosphorylation, methylation, acetylation, ubiquitination, SUMOylation, and O-GlcNAcylation. This review focuses on our current understanding and the emerging evidence of how post-translational regulations modify transcription factors involved in the etiology and pathophysiology of colonic disease as well as the implications of these findings for new therapeutic approaches in these disorders.
Collapse
Affiliation(s)
- Chao-Yuan Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, No.35, Keyan Road, Zhunan, Miaoli 350, Taiwan; (C.-Y.H.); (S.-H.F.)
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, No.161, Section 6, Min Chuan East Road, Neihu, Taipei 114, Taiwan; (M.-W.C.); (S.-J.C.)
| | - Shin-Huei Fu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, No.35, Keyan Road, Zhunan, Miaoli 350, Taiwan; (C.-Y.H.); (S.-H.F.)
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, No.161, Section 6, Min Chuan East Road, Neihu, Taipei 114, Taiwan; (M.-W.C.); (S.-J.C.)
| | - Ming-Wei Chien
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, No.161, Section 6, Min Chuan East Road, Neihu, Taipei 114, Taiwan; (M.-W.C.); (S.-J.C.)
| | - Yu-Wen Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, No.161, Section 6, Min Chuan East Road, Neihu, Taipei 114, Taiwan;
- Molecular Cell Biology, Taiwan International Graduate Program, No.128, Academia Road, Section 2, Nankang, Taipei 115, Taiwan
| | - Shyi-Jou Chen
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, No.161, Section 6, Min Chuan East Road, Neihu, Taipei 114, Taiwan; (M.-W.C.); (S.-J.C.)
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Chenggong Rd., Neihu District, Taipei 114, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, No.35, Keyan Road, Zhunan, Miaoli 350, Taiwan; (C.-Y.H.); (S.-H.F.)
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, No.161, Section 6, Min Chuan East Road, Neihu, Taipei 114, Taiwan; (M.-W.C.); (S.-J.C.)
- Graduate Institute of Life Sciences, National Defense Medical Center, No.161, Section 6, Min Chuan East Road, Neihu, Taipei 114, Taiwan;
- Correspondence: ; Tel.: +886-2-8792-3100 (ext. 18539); Fax: +886-2-8792-1774
| |
Collapse
|
11
|
Gu ZL, Huang J, Zhen LL. Knockdown of otubain 2 inhibits liver cancer cell growth by suppressing NF-κB signaling. Kaohsiung J Med Sci 2020; 36:399-404. [PMID: 32003539 DOI: 10.1002/kjm2.12187] [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] [Received: 11/12/2019] [Accepted: 01/07/2020] [Indexed: 12/13/2022] Open
Abstract
The deubiquitinase otubain 2 (OTUB2) has been reported to play significant roles in the tumorigenesis of several cancers, but the role of OTUB2 in liver cancer is not investigated yet. In the present study, OTUB2 was found significantly upregulated in liver cancer tumor tissues and cell lines, and elevated OTUB2 indicated as a negative index for the overall survival of liver cancer patients. At the cellular level, knockdown of OTUB2 markedly inhibited liver cancer cell growth. Our further investigations revealed that knockdown of OTUB2 significantly suppressed NF-κB-driving luciferase activity, and markedly inhibited the phosphorylation of NF-κB p65 in liver cancer cells, which indicated that OTUB2 mediated liver cancer cell growth by regulating NF-κB signaling. Additionally, we found that liver cancer cell lines harboring higher OTUB2 expression were more sensitive to NF-κB inhibitors, and overexpression of OTUB2 could significantly reduce the antitumor effects of NF-κB inhibitors in liver cancer cells. This study indicated that OTUB2 could be a promising target for the treatment of liver cancer in the future.
Collapse
Affiliation(s)
- Zhen-Lin Gu
- Department of Interventional Radiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Jing Huang
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Lin-Lin Zhen
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| |
Collapse
|
12
|
Xu X, Li S, Cui X, Han K, Wang J, Hou X, Cui L, He S, Xiao J, Yang Y. Inhibition of Ubiquitin Specific Protease 1 Sensitizes Colorectal Cancer Cells to DNA-Damaging Chemotherapeutics. Front Oncol 2019; 9:1406. [PMID: 31921663 PMCID: PMC6930197 DOI: 10.3389/fonc.2019.01406] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022] Open
Abstract
Mutations and altered expression of deubiquitinating enzymes (DUBs) have been found associated with many human diseases including cancers. In this study, Ubiquitin specific protease 1 (USP1) expression was found significantly increased in some colorectal cancers (CRC). The elevated USP1 level was associated with short overall survival of patients and with advanced stages of cancers. In cultured CRC cells, knockdown of USP1 induced growth arrest at G2/M of cell cycle and reduced the expression of anti-apoptotic proteins Bcl-2 and Mcl-1. Its knockdown also led to reduction of DNA-repair related substrates FANCD2 and ID1. Further investigations found that small molecular inhibitor of USP1 ML323 sensitized CRC cells to DNA-targeting chemotherapeutics, including doxorubicin, TOPI/II inhibitors, and PARP inhibitor, but not to 5-Fu. These results indicate that USP1 plays a critical in colorectal cancer cell survival and is a promising target for anti-colorectal cancer chemotherapy. Targeting USP1 may represent an effective strategy to regulate the DNA-repairing system.
Collapse
Affiliation(s)
- Xin Xu
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Shaoyan Li
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ximao Cui
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Colorectal Cancer Research Center, Shanghai, China
| | - Kunkun Han
- The Asclepius Technology Company Group and Asclepius Cancer Research Center, Suzhou, China
| | - Jun Wang
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaodan Hou
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Long Cui
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Colorectal Cancer Research Center, Shanghai, China
| | - Songbing He
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiecheng Xiao
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yili Yang
- Center for Systems Medicine, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,State Key Laboratory of Innovative Natural Medicine and TCM Injections, Qingfeng Pharmaceutical, Ganzhou, China
| |
Collapse
|
13
|
He Y, Wang S, Tong J, Jiang S, Yang Y, Zhang Z, Xu Y, Zeng Y, Cao B, Moran MF, Mao X. The deubiquitinase USP7 stabilizes Maf proteins to promote myeloma cell survival. J Biol Chem 2019; 295:2084-2096. [PMID: 31822558 DOI: 10.1074/jbc.ra119.010724] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/25/2019] [Indexed: 12/11/2022] Open
Abstract
The Maf proteins, including c-Maf, MafA, and MafB, are critical transcription factors in myelomagenesis. Previous studies demonstrated that Maf proteins are processed by the ubiquitin-proteasome pathway, but the mechanisms remain elusive. This study applied MS to identify MafB ubiquitination-associated proteins and found that the ubiquitin-specific protease USP7 was present in the MafB interactome. Moreover, USP7 also interacted with c-Maf and MafA and blocked their polyubiquitination and degradation. Consistently, knockdown of USP7 resulted in Maf protein degradation along with increased polyubiquitination levels. The action of USP7 thus promoted Maf transcriptional activity as evidenced by luciferase assays and by the up-regulation of the expression of Maf-modulated genes. Furthermore, USP7 was up-regulated in myeloma cells, and it was negatively associated with the survival of myeloma patients. USP7 promoted myeloma cell survival, and when it was inhibited by its specific inhibitor P5091, myeloma cell lines underwent apoptosis. These results therefore demonstrated that USP7 is a deubiquitinase of Maf proteins and promotes MM cell survival in association with Maf stability. Given the significance of USP7 and Maf proteins in myeloma genesis, targeting the USP7/Maf axle is a potential strategy to the precision therapy of MM.
Collapse
Affiliation(s)
- Yuanming He
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; Guangzhou and Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou 511436, China
| | - Siyu Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jiefei Tong
- Program in Cell Biology, Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto M5G 0A4, Canada
| | - Shuoyi Jiang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; Guangzhou and Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou 511436, China
| | - Ye Yang
- School of Basic Medicine, Nanjing University of Traditional Medicine, Nanjing 210023, China
| | - Zubin Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yujia Xu
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yuanying Zeng
- Department of Oncology, Suzhou Municipal Hospital, Suzhou 215100, China.
| | - Biyin Cao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Michael F Moran
- Program in Cell Biology, Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto M5G 0A4, Canada
| | - Xinliang Mao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; Guangzhou and Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou 511436, China; Guangzhou Institute of Cardiovascular Disease and Department of Hematology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
14
|
Mebendazole elicits potent antimyeloma activity by inhibiting the USP5/c-Maf axis. Acta Pharmacol Sin 2019; 40:1568-1577. [PMID: 31197245 PMCID: PMC7468578 DOI: 10.1038/s41401-019-0249-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/08/2019] [Indexed: 01/22/2023]
Abstract
c-Maf is a critical oncogenic transcription factor that contributes to myelomagenesis. Our previous studies demonstrated that the deubiquitinase USP5 stabilizes c-Maf and promotes myeloma cell proliferation and survival; therefore, the USP5/c-Maf axis could be a potential target for myeloma therapy. As a concept of principle, the present study established a USP5/c-Maf-based luciferase system that was used to screen an FDA-approved drug library. It was found that mebendazole, a typical anthelmintic drug, preferentially induced apoptosis in c-Maf-expressing myeloma cells. Moreover, oral administration of mebendazole delayed the growth of human myeloma xenografts in nude mice but did not show overt toxicity. Further studies showed that the selective antimyeloma activity of mebendazole was associated with the inhibition of the USP5/c-Maf axis. Mebendazole downregulated USP5 expression and disrupted the interaction between USP5 and c-Maf, thus leading to increased levels of c-Maf ubiquitination and subsequent c-Maf degradation. Mebendazole inhibited c-Maf transcriptional activity, as confirmed by both luciferase assays and expression measurements of c-Maf downstream genes. In summary, this study identified mebendazole as a USP5/c-Maf inhibitor that could be developed as a novel antimyeloma agent.
Collapse
|
15
|
Zhang YK, Tian WZ, Zhang RS, Zhang YJ, Ma HT. Ubiquitin-specific protease 44 inhibits cell growth by suppressing AKT signaling in non-small cell lung cancer. Kaohsiung J Med Sci 2019; 35:535-541. [PMID: 31197957 DOI: 10.1002/kjm2.12096] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/14/2019] [Indexed: 12/24/2022] Open
Abstract
Ubiquitin-specific protease 44 (USP44) has been reported as a tumor suppressor or promoter in some tumors, but its function in non-small cell lung cancer (NSCLC) is still unclear. In this study, USP44 was found significantly downregulated in both of NSCLC tissues and cell lines, and low expression of USP44 predicted a poor prognosis for NSCLC patients. Overexpression of USP44 markedly downregulated the expression levels of Cyclin D1 and CDK4, but upregulated p53 expression, as a result of which, suppressing the cell growth of NSCLC cells. Further studies indicated that overexpression of USP44 significantly inhibited the phosphorylation of AKT, and its down-stream signals, including mTOR and P70S6K. Moreover, overexpression of USP44 increased PTEN protein but not its mRNA levels, which suggested that USP44 inhibited AKT signaling by stabilizing PTEN in NSCLC cells. In conclusion, we demonstrated that USP44 showed prior evidence of a tumor suppressive function in NSCLC cells, and inhibited NSCLC cell growth by suppressing AKT signaling, suggesting that USP44 could be as a novel target for NSCLC therapy.
Collapse
Affiliation(s)
- Yun-Kui Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Thoracic Surgery, Shanxi Tumor Hospital, Taiyuan, China
| | - Wen-Ze Tian
- Department of Thoracic Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Rong-Sheng Zhang
- Department of Thoracic Surgery, Shanxi Tumor Hospital, Taiyuan, China
| | - Yu-Jie Zhang
- Department of Thoracic Surgery, Shanxi Tumor Hospital, Taiyuan, China
| | - Hai-Tao Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
16
|
Xu X, Huang A, Cui X, Han K, Hou X, Wang Q, Cui L, Yang Y. Ubiquitin specific peptidase 5 regulates colorectal cancer cell growth by stabilizing Tu translation elongation factor. Theranostics 2019; 9:4208-4220. [PMID: 31281542 PMCID: PMC6592179 DOI: 10.7150/thno.33803] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Ubiquitin specific peptidase 5 (USP5) is a ubiquitous expressed deubiquitinating enzyme (DUB). It has been shown involved in DNA repair, apoptosis, inflammation, and tumor cell growth. However, the function and molecular mechanism of USP5 in colorectal cancer (CRC) are still unclear. In the present study, we asked how it affected the growth of colorectal cancer cells. Methods: A shRNA-based high-content screening was performed to identify DUBs affecting the growth of CRC cells. CCK-8 assay and xenografts were used to assess CRC cell growth, survival and tumorigenesis. RT-qPCR, immunoblotting and immunohistochemistry were carried out to quantitate USP5 expression in CRC tissues and cell lines. Immunoprecipitation and mass spectrometry analysis were performed to identify USP5-interacting proteins. Cycloheximide chase was performed to assess Tu translation elongation factor (TUFM) stability. Dual luciferase reporter assay was utilized for USP5 promoter analysis. Results: We found that USP5 was highly expressed in a group of primary CRC tissues, and the increased USP5 was correlated with clinical stages and shorter overall survival. While USP5 knockdown effectively inhibited CRC cell growth, overexpressed USP5 promoted the growth of CRC cells and made them more resistant to doxorubicin (DOX). TUFM was discovered as a substrate of USP5. USP5 deubiquitinated TUFM and increased its level in CRC cells. Enforced expression of TUFM was able to alleviate the growth inhibition induced by USP5 knockdown. Further analyses showed that EBF transcription factor 1 (EBF1) was a major regulator for USP5 transcription, and DOX inhibited EBF1-USP5-TUFM axis in CRC cells. Conclusions: USP5 was required for CRC cells and promoted their growth and resistance to chemotherapeutics. TUFM was a USP5 deubiquitinating substrate that mediated the cellular effects of USP5. The transcription of USP5 was regulated by EBF1. Thus, targeting EBF1-USP5-TUFM axis is a potential novel strategy for CRC treatment.
Collapse
|
17
|
Huang Z, Ge H, Yang C, Cai Y, Chen Z, Tian W, Tao J. MicroRNA‐26a‐5p inhibits breast cancer cell growth by suppressing RNF6 expression. Kaohsiung J Med Sci 2019; 35:467-473. [PMID: 31063232 DOI: 10.1002/kjm2.12085] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/22/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Zi‐Ming Huang
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Heng‐Fa Ge
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Chen‐Chen Yang
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Yong Cai
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Zhen Chen
- Department of Emergency SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Wen‐Ze Tian
- Department of Cardio‐Thoracic SurgeryThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| | - Jia‐Li Tao
- Department of EmergencyThe Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University Huai'an Jiangsu China
| |
Collapse
|
18
|
Shen WM, Yin JN, Xu RJ, Xu DF, Zheng SY. Ubiquitin specific peptidase 49 inhibits non-small cell lung cancer cell growth by suppressing PI3K/AKT signaling. Kaohsiung J Med Sci 2019; 35:401-407. [PMID: 31001918 DOI: 10.1002/kjm2.12073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/01/2019] [Indexed: 12/16/2022] Open
Abstract
Ubiquitin specific peptidase 49 (USP49) has been reported as a tumor suppressor in several tumors, but its function and molecular mechanism in non-small cell lung cancer (NSCLC) are still unknown. In this study, USP49 was found downregulated in NSCLC primary tissues and cell lines, and high USP49 predicted a positive index for the overall survival of NSCLC patients. Overexpression of USP49 downregulated the expression levels of Cyclin D1, and upregulated p53 expression. Further flow cytometry analysis showed that overexpressed USP49 induced cell cycle arrest at G0/G1 phase. As a result, overexpression of USP49 significantly inhibited cell growth of NSCLC cells. In mechanism, overexpression of USP49 inhibited PI3K/AKT signaling, but knockdown of USP49 enhanced this signaling. Further studies indicated that USP49 deubiquitinated PTEN and stabilized PTEN protein, which suggested that USP49 inhibited PI3K/AKT signaling by stabilizing PTEN in NSCLC cells. In conclusion, we demonstrated that USP49 was functional in NSCLC cells, and inhibited NSCLC cell growth by suppressing PI3K/AKT signaling, suggesting that USP49 could be as a novel target for NSCLC therapy.
Collapse
Affiliation(s)
- Wen-Ming Shen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Emergency Surgery, The Affiliated Wujin People's Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Jin-Nan Yin
- Department of Emergency Surgery, The Affiliated Wujin People's Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Rui-Jun Xu
- Department of Endocrinology, The Affiliated Wujin People's Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Da-Fu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Thoracic Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Shi-Ying Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
19
|
Li P, Huang Z, Wang J, Chen W, Huang J. Ubiquitin-specific peptidase 28 enhances STAT3 signaling and promotes cell growth in non-small-cell lung cancer. Onco Targets Ther 2019; 12:1603-1611. [PMID: 30881015 PMCID: PMC6396656 DOI: 10.2147/ott.s194917] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Ubiquitin-specific peptidase 28 (USP28) has been reported to play significant roles in several tumors, but its roles in non-small-cell lung cancer (NSCLC) is still unknown. In this study, we aimed to investigate the biological function and molecular mechanisms of USP28 in NSCLC. MATERIALS AND METHODS Immunoblotting analysis was used to detect relative proteins' expression. Luciferase assay was performed to explore the activation of signal transducer and activator of transcription 3 (STAT3). Immunoprecipitation was performed to assess whether USP28 interacted with STAT3 or deubiquitinated STAT3. Quantitative real-time PCR was performed to evaluate the relative mRNA levels of STAT3 and USP28. Cycloheximide chase assay was carried out to examine whether USP28 affected the half-life of STAT3 protein. Cell Counting Kit-8 assay and xenograft model were used to assess whether USP28 regulated NSCLC cell growth. RESULTS In this study, the deubiquitinating enzyme USP28 was found to mediate STAT3 signaling in NSCLC cells. USP28 interacted with STAT3, and increased the stability of STAT3 by inducing its deubiquitination. Further studies showed that USP28 was upregulated in both the primary tissues and cell lines of NSCLC. The Kaplan-Meier plotter also indicated that USP28 predicted a poor prognosis of NSCLC patients. Moreover, knockdown of USP28 inhibited cell growth of NSCLC cells in vitro and delayed NSCLC tumor growth in vivo. CONCLUSION These results demonstrated that USP28 was functional in NSCLC cells, and promoted NSCLC cell growth by inducing STAT3 signaling. This suggests that USP28 could be a novel target for NSCLC therapy.
Collapse
Affiliation(s)
- Pengling Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China,
- Department of Respiratory Medicine, The Affiliated Huai'an No 1. People's Hospital, Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Ziming Huang
- Department of Emergency Surgery, The Affiliated Huai'an No 1. People's Hospital, Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Jipeng Wang
- Department of Respiratory Medicine, The Affiliated Huai'an No 1. People's Hospital, Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Wei Chen
- Department of Respiratory Medicine, The Affiliated Huai'an No 1. People's Hospital, Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China,
| |
Collapse
|
20
|
Xu D, Tian W, Jiang C, Huang Z, Zheng S. The anthelmintic agent oxfendazole inhibits cell growth in non‑small cell lung cancer by suppressing c‑Src activation. Mol Med Rep 2019; 19:2921-2926. [PMID: 30720086 DOI: 10.3892/mmr.2019.9897] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 12/04/2018] [Indexed: 11/05/2022] Open
Abstract
The c‑Src protein family of tyrosine kinases are important in the tumorigenesis of many types of tumors, and may be a potential target for antitumor drug discovery. In the present study, immunoblotting was performed to analyze protein expression, CCK‑8 assay was carried out to assess cell viability and cell cycle was analyzed using a flow cytometer. The anthelmintic agent oxfendazole was observed to be a novel c‑Src inhibitor that blocked the activation of c‑Src. Oxfendazole also suppressed the cell growth of non‑small cell lung cancer (NSCLC) cells, and overexpression of c‑Src decreased the cytotoxicity of oxfendazole against NSCLC cells. In addition, oxfendazole induced cell cycle arrest at the G0/G1 phase, and downregulated the protein levels of Cyclin‑dependent kinase (CDK)‑4, CDK6, retinoblastoma protein and E2 transcription factor 1, and upregulated the expression levels of p53 and p21 in NSCLC cells. Furthermore, oxfendazole enhanced the cytotoxicity of cisplatin against NSCLC cells. These results demonstrated that oxfendazole exerted its antitumor activity by suppressing c‑Src signaling, and it was also indicated that the anthelmintic agent oxfendazole may be effective for anti‑NSCLC therapy in the clinic as a single agent or in combination with other antitumor drugs.
Collapse
Affiliation(s)
- Dafu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wenze Tian
- Department of Thoracic Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Chao Jiang
- Department of Oncology, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Ziming Huang
- Department of Emergency Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Shiying Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
21
|
Tao J, Zhao H, Xie X, Luo M, Gao Z, Sun H, Huang Z. The anthelmintic drug flubendazole induces cell apoptosis and inhibits NF-κB signaling in esophageal squamous cell carcinoma. Onco Targets Ther 2019; 12:471-478. [PMID: 30666126 PMCID: PMC6331185 DOI: 10.2147/ott.s193206] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The nuclear factor kappa B (NF-κB) signaling is activated in esophageal squamous cell carcinoma (ESCC) and can be used as a potential target for anti-ESCC drug discovery. In this study, we aimed to investigate the function of flubendazole as a novel NF-κB inhibitor in ESCC cells. MATERIALS AND METHODS Cell Counting Kit-8 assay was carried out to assess cell viability of ESCC cells. Flow cytometry and immunoblotting were performed to examine cell apoptosis. Immunoblotting assay was used to analyze the protein expression of NF-κB signaling. Luciferase assay was performed to explore the activation of NF-κB. Plasmids were transfected into ESCC cells using Lipofectamine® 2000. RESULTS In this study, the anthelmintic drug flubendazole was found to inhibit the activation of IκBα kinases (IKKs), block the activation of IκBα, and decrease the phosphorylation of NF-κB p65, which could be a novel NF-κB inhibitor in ESCC cells. We also found that flubendazole inhibited the cell survival of different ESCC cells and induced cell apoptosis in both EC9706 and TE1 cells. Moreover, overexpression of constitutively activated IKKβ markedly decreased the cytotoxic effect of flubendazole on EC9706 and TE1 cells. In addition, flubendazole also showed a synergistic effect on ESCC cells when combined with doxorubicin. CONCLUSION The results above demonstrated that flubendazole showed its anti-tumor action by suppressing the NF-κB signaling pathway and suggested that flubendazole might be re-purposed for anti-ESCC therapy in clinic as a single agent or in combination with other anti-tumor drugs.
Collapse
Affiliation(s)
- Jiali Tao
- Department of Emergency, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Hongmei Zhao
- Department of Emergency, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Xiaochen Xie
- Department of Respiratory, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Man Luo
- Department of Emergency, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Zhiwei Gao
- Department of Emergency, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Hong Sun
- Department of Emergency, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Ziming Huang
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| |
Collapse
|
22
|
Liu Y, Wang X, Zeng S, Zhang X, Zhao J, Zhang X, Chen X, Yang W, Yang Y, Dong Z, Zhu J, Xu X, Tian F. The natural polyphenol curcumin induces apoptosis by suppressing STAT3 signaling in esophageal squamous cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:303. [PMID: 30518397 PMCID: PMC6280482 DOI: 10.1186/s13046-018-0959-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022]
Abstract
Background We and others have previously shown that the STAT3 signaling pathway is activated in some esophageal squamous cell carcinoma (ESCC) cells and is required for the survival and growth of these primary ESCC-derived xenografts. It has also been shown that the natural polyphenol curcumin is an effective anti-tumor agent. Methods Luciferase assay and immunoblotting were performed to examine whether curcumin suppressed STAT3 signaling. CCK-8 assay and xenografts were utilized for analyzing ESCC cell growth in culture and mice. Soft agar assay was carried out to determine the colony formation ability of ESCC cells in the presence or absence of curcumin. Cell death and cell cycle were assessed by In CELL Analyzer 2000. Immunohistochemistry and TUNEL assay were used for detecting apoptosis in ESCC tisuses. Molecular docking was performed to evaluate the interaction of curcumin with JAK2. JAK2 activity was assessed using an in vitro cell-free system. HE staining was used to evaluate the ESCC tissues. Results The natural polyphenol curcumin inhibited STAT3 phosphorylation rapidly and blocked STAT3-mediated signaling in ESCC cells. It also induced growth arrest and apoptosis in cultured ESCC cells, which were attenuated by enforced expression of STAT3. Furthermore, curcumin preferentially blocked the growth of primary ESCC-derived xenografts that harbored activated STAT3. Conclusions Curcumin is able to exert anti-tumor action through inhibiting the STAT3 signaling pathway. Giving its wide use in traditional medicines with low toxicity and few adverse reactions, it is conceivable that curcumin might be further explored as a unique STAT3 inhibitor for anti-cancer therapies.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China.,Clinical Research Center, People's Hospital of Zhengzhou, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xinhua Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China
| | - Shuang Zeng
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xiane Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xiaoyan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Wanjing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Yili Yang
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, 215123, People's Republic of China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China
| | - Jingyu Zhu
- School of Medicine and Pharmaceutics, Jiangnan University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Xin Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China. .,Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, 215123, People's Republic of China.
| | - Fang Tian
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China. .,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, 450001, People's Republic of China.
| |
Collapse
|
23
|
Huang Z, Cai Y, Yang C, Chen Z, Sun H, Xu Y, Chen W, Xu D, Tian W, Wang H. Knockdown of RNF6 inhibits gastric cancer cell growth by suppressing STAT3 signaling. Onco Targets Ther 2018; 11:6579-6587. [PMID: 30323630 PMCID: PMC6178940 DOI: 10.2147/ott.s174846] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background and objective RNF6, an E3 ligase, has been reported to play an important role in the tumorigenesis in several tissues, but its role in gastric cancer is still unknown. In this study, we aimed to investigate the biological function and molecular mechanisms of RNF6 in gastric cancer. Materials and methods The expression levels of RNF6 were detected by quantitative real-time PCR (qRT-PCR) and immunoblotting in gastric cancer tissues and cell lines. Cell Counting Kit-8 assay was performed to evaluate cell proliferation. Cell apoptosis was analyzed by flow cytometer and immunoblotting. Luciferase assay, immunoblotting and qRT-PCR were performed to explore the activation of STAT3. Immunoprecipitation was performed to evaluate the ubiquitination of SHP-1. Results In this study, RNF6 was found to be upregulated in both primary tissues and cell lines of gastric cancer. Knockdown or overexpression of RNF6 inhibited or promoted cell growth of gastric cancer cells. Knockdown of RNF6 also induced the cleavage of PARP and promoted cell apoptosis in gastric cancer cells. In addition, knockdown of RNF6 also increased the cytotoxicity of doxorubicin against gastric cancer. Moreover, knockdown of RNF6 inhibited STAT3-derived luciferase activity and downregulated the phosphorylation of STAT3, but upregulated the protein level of SHP-1. Knockdown of RNF6 downregulated the expression of MCL1 and XIAP, which are target genes of STAT3. Further studies showed that RNF6 regulated the stability of SHP-1 by inducing its polyubiquitination. Conclusion These results demonstrated that RNF6 was highly expressed in gastric cancer and regulated the growth of gastric cancer cells by affecting SHP-1/STAT3 signaling, which suggested that RNF6 could be a novel target for gastric cancer therapy.
Collapse
Affiliation(s)
- Ziming Huang
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Yong Cai
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Chenchen Yang
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Zhen Chen
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Hong Sun
- Department of Emergency Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| | - Yingying Xu
- Department of Radiotherapy, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Wei Chen
- Department of Respiratory Care, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Dafu Xu
- Department of Cardiothoracic Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Wenze Tian
- Department of Cardiothoracic Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Haixiao Wang
- Department of General Surgery, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China,
| |
Collapse
|
24
|
Zhang J, Wu H, Yi B, Zhou J, Wei L, Chen Y, Zhang L. RING finger protein 38 induces gastric cancer cell growth by decreasing the stability of the protein tyrosine phosphatase SHP-1. FEBS Lett 2018; 592:3092-3100. [PMID: 30112836 DOI: 10.1002/1873-3468.13225] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/31/2018] [Accepted: 08/08/2018] [Indexed: 12/15/2022]
Abstract
The function of the E3 ligase RNF38 is still unknown in gastric cancer. Here, we found that RNF38 is upregulated in gastric cancer, and it is associated with the overall survival of gastric cancer patients. Further studies showed that RNF38 interacts with the nonreceptor tyrosine phosphatase SH2-containing protein tyrosine phosphatase 1 (SHP-1) and induces the polyubiquitination of SHP-1, which leads to destabilization of SHP-1 and promotion of STAT3 signaling in gastric cancer cells. In addition, overexpression or knockdown of RNF38 induces or suppresses gastric cancer cell growth in vitro, respectively, and silencing RNF38 delays tumor growth in vivo. These findings demonstrate that RNF38 is functional in gastric cancer and promotes STAT3 signaling by destabilizing SHP-1; thus, RNF38 could be a novel target for gastric cancer therapy.
Collapse
Affiliation(s)
- Jigang Zhang
- Department of Emergency Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Wu
- Department of Emergency Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Yi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Luxin Wei
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lifeng Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
25
|
Hsu CY, Yeh LT, Fu SH, Chien MW, Liu YW, Miaw SC, Chang DM, Sytwu HK. SUMO-defective c-Maf preferentially transactivates Il21 to exacerbate autoimmune diabetes. J Clin Invest 2018; 128:3779-3793. [PMID: 30059018 DOI: 10.1172/jci98786] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 06/14/2018] [Indexed: 12/19/2022] Open
Abstract
SUMOylation is involved in the development of several inflammatory diseases, but the physiological significance of SUMO-modulated c-Maf in autoimmune diabetes is not completely understood. Here, we report that an age-dependent attenuation of c-Maf SUMOylation in CD4+ T cells is positively correlated with the IL-21-mediated diabetogenesis in NOD mice. Using 2 strains of T cell-specific transgenic NOD mice overexpressing wild-type c-Maf (Tg-WTc) or SUMOylation site-mutated c-Maf (Tg-KRc), we demonstrated that Tg-KRc mice developed diabetes more rapidly than Tg-WTc mice in a CD4+ T cell-autonomous manner. Moreover, SUMO-defective c-Maf preferentially transactivated Il21 to promote the development of CD4+ T cells with an extrafollicular helper T cell phenotype and expand the numbers of granzyme B-producing effector/memory CD8+ T cells. Furthermore, SUMO-defective c-Maf selectively inhibited recruitment of Daxx/HDAC2 to the Il21 promoter and enhanced histone acetylation mediated by CREB-binding protein (CBP) and p300. Using pharmacological interference with CBP/p300, we illustrated that CBP30 treatment ameliorated c-Maf-mediated/IL-21-based diabetogenesis. Taken together, our results show that the SUMOylation status of c-Maf has a stronger regulatory effect on IL-21 than the level of c-Maf expression, through an epigenetic mechanism. These findings provide new insights into how SUMOylation modulates the pathogenesis of autoimmune diabetes in a T cell-restricted manner and on the basis of a single transcription factor.
Collapse
Affiliation(s)
| | - Li-Tzu Yeh
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Shin-Huei Fu
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Wei Chien
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Wen Liu
- Graduate Institute of Life Sciences and.,Molecular Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
| | - Shi-Chuen Miaw
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Deh-Ming Chang
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Huey-Kang Sytwu
- Graduate Institute of Life Sciences and.,Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
26
|
Du Y, Liu Y, Xu Y, Juan J, Zhang Z, Xu Z, Cao B, Wang Q, Zeng Y, Mao X. The transmembrane protein TMEPAI induces myeloma cell apoptosis by promoting degradation of the c-Maf transcription factor. J Biol Chem 2018; 293:5847-5859. [PMID: 29467225 DOI: 10.1074/jbc.ra117.000972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/14/2018] [Indexed: 11/06/2022] Open
Abstract
TMEPAI (transmembrane prostate androgen-induced protein, also called prostate transmembrane protein, androgen-induced 1 (PMEPA1)) is a type I transmembrane (TM) protein, but its cellular function is largely unknown. Here, studying factors influencing the stability of c-Maf, a critical transcription factor in multiple myeloma (MM), we found that TMEPAI induced c-Maf degradation. We observed that TMEPAI recruited NEDD4 (neural precursor cell expressed, developmentally down-regulated 4), a WW domain-containing ubiquitin ligase, to c-Maf, leading to its degradation through the proteasomal pathway. Further investigation revealed that TMEPAI interacts with NEDD4 via its conserved PY motifs. Alanine substitution or deletion of these motifs abrogated the TMEPAI complex formation with NEDD4, resulting in failed c-Maf degradation. Functionally, TMEPAI suppressed the transcriptional activity of c-Maf. Of note, increased TMEPAI expression was positively associated with the overall survival of MM patients. Moreover, TMEPAI was down-regulated in MM cells, and re-expression of TMEPAI induced MM cell apoptosis. In conclusion, this study highlights that TMEPAI decreases c-Maf stability by recruiting the ubiquitin ligase NEDD4 to c-Maf for proteasomal degradation. Our findings suggest that the restoration of functional TMEPA1 expression may represent a promising complementary therapeutic strategy for treating patients with MM.
Collapse
Affiliation(s)
- Yanyun Du
- From the Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yan Liu
- From the Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yujia Xu
- From the Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jiaxiang Juan
- From the Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zubin Zhang
- From the Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhuan Xu
- the Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou 215100, China
| | - Biyin Cao
- From the Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qi Wang
- the Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yuanying Zeng
- Department of Oncology, Suzhou Municipal Hospital, Suzhou 215100, China, and
| | - Xinliang Mao
- From the Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China, .,the Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
27
|
Inhibition of the deubiquitinase USP5 leads to c-Maf protein degradation and myeloma cell apoptosis. Cell Death Dis 2017; 8:e3058. [PMID: 28933784 PMCID: PMC5636991 DOI: 10.1038/cddis.2017.450] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 12/04/2022]
Abstract
The deubiquitinase USP5 stabilizes c-Maf, a key transcription factor in multiple myeloma (MM), but the mechanisms and significance are unclear. In the present study, USP5 was found to interact with c-Maf and prevented it from degradation by decreasing its polyubiquitination level. Specifically, the 308th and 347th lysine residues in c-Maf were critical for USP5-mediated deubiquitination and stability. There are five key domains in the USP5 protein and subsequent studies revealed that the cryptic ZnF domain and the C-box domain interacted with c-Maf but the UBA1/UBA2 domain partly increased its stability. Notably, MafA and MafB are also members of the c-Maf family, however, USP5 failed to deubiquitinate MafA, suggesting its substrate specificity. In the functional studies, USP5 was found to promoted the transcriptional activity of c-Maf. Consistent with the high level of c-Maf protein in MM cells, USP5 was also highly expressed. When USP5 was knocked down, c-Maf underwent degradation. Interestingly, USP5 silence led to apoptosis of MM cells expressing c-Maf but not MM cells lacking c-Maf, indicating c-Maf is a key factor in USP5-mediated MM cell proliferation and survival. Consistent with this finding, WP1130, an inhibitor of several Dubs including USP5, suppressed the transcriptional activity of c-Maf and induced MM cell apoptosis. When c-Maf was overexpressed, WP1130-induced MM cell apoptosis was abolished. Taken together, these findings suggest that USP5 regulates c-Maf stability and MM cell survival. Targeting the USP5/c-Maf axis could be a potential strategy for MM treatment.
Collapse
|
28
|
Xu Y, Zhang Z, Li J, Tong J, Cao B, Taylor P, Tang X, Wu D, Moran MF, Zeng Y, Mao X. The ubiquitin-conjugating enzyme UBE2O modulates c-Maf stability and induces myeloma cell apoptosis. J Hematol Oncol 2017; 10:132. [PMID: 28673317 PMCID: PMC5496436 DOI: 10.1186/s13045-017-0499-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/16/2017] [Indexed: 12/31/2022] Open
Abstract
Background UBE2O is proposed as a ubiquitin-conjugating enzyme, but its function was largely unknown. Methods Mass spectrometry was applied to identify c-Maf ubiquitination-associated proteins. Immunoprecipitation was applied for c-Maf and UBE2O interaction. Immunoblotting was used for Maf protein stability. Luciferase assay was used for c-Maf transcriptional activity. Lentiviral infections were applied for UBE2O function in multiple myeloma (MM) cells. Flow cytometry and nude mice xenografts were applied for MM cell apoptosis and tumor growth assay, respectively. Results UBE2O was found to interact with c-Maf, a critical transcription factor in MM, by the affinity purification/tandem mass spectrometry assay and co-immunoprecipitation assays. Subsequent studies showed that UBE2O mediated c-Maf polyubiquitination and degradation. Moreover, UBE2O downregulated the transcriptional activity of c-Maf and the expression of cyclin D2, a typical gene modulated by c-Maf. DNA microarray revealed that UBE2O was expressed in normal bone marrow cells but downregulated in MGUS, smoldering MM and MM cells, which was confirmed by RT-PCR in primary MM cells, suggesting its potential role in myeloma pathophysiology. When UBE2O was restored, c-Maf protein in MM cells was significantly decreased and MM cells underwent apoptosis. Furthermore, the human MM xenograft in nude mice showed that re-expression of UBE2O delayed the growth of myeloma xenografts in nude mice in association with c-Maf downregulation and activation of the apoptotic pathway. Conclusions UBE2O mediates c-Maf polyubiquitination and degradation, induces MM cell apoptosis, and suppresses myeloma tumor growth, which provides a novel insight in understanding myelomagenesis and UBE2O biology.
Collapse
Affiliation(s)
- Yujia Xu
- Jiangsu Key Laboratory for Translational Research and Therapeutics of Neuro-Psycho- Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Zubin Zhang
- Jiangsu Key Laboratory for Translational Research and Therapeutics of Neuro-Psycho- Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Jie Li
- Jiangsu Key Laboratory for Translational Research and Therapeutics of Neuro-Psycho- Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Jiefei Tong
- Program in Molecular Structure and Function, The Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, M5G 0A4, Canada
| | - Biyin Cao
- Jiangsu Key Laboratory for Translational Research and Therapeutics of Neuro-Psycho- Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Paul Taylor
- Program in Molecular Structure and Function, The Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, M5G 0A4, Canada
| | - Xiaowen Tang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Michael F Moran
- Program in Molecular Structure and Function, The Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, M5G 0A4, Canada
| | - Yuanying Zeng
- Jiangsu Key Laboratory for Translational Research and Therapeutics of Neuro-Psycho- Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China. .,Department of Oncology, Suzhou Municipal Hospital East Campus, Suzhou, 215100, People's Republic of China.
| | - Xinliang Mao
- Jiangsu Key Laboratory for Translational Research and Therapeutics of Neuro-Psycho- Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China. .,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
29
|
The ubiquitin ligase HERC4 mediates c-Maf ubiquitination and delays the growth of multiple myeloma xenografts in nude mice. Blood 2016; 127:1676-86. [PMID: 26825710 DOI: 10.1182/blood-2015-07-658203] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 12/18/2015] [Indexed: 01/11/2023] Open
Abstract
The transcription factor c-Maf is extensively involved in the pathophysiology of multiple myeloma (MM), a fatal malignancy of plasma cells. In the present study, affinity chromatography and mass spectrometry were used to identify c-Maf ubiquitination-associated proteins, from which the E3 ligase HERC4 was found to interact with c-Maf and catalyzed its polyubiquitination and subsequent proteasome-mediated degradation. HERC4 mediated polyubiquitination at K85 and K297 in c-Maf, and this polyubiquitination could be prevented by the isopeptidase USP5. Further analysis on the NCI-60 cell line collection revealed that RPMI 8226, a MM-derived cell line, expressed the lowest level of HERC4. Primary bone marrow analysis revealed HERC4 expression was high in normal bone marrow, but was steadily decreased during myelomagenesis. These findings suggested HERC4 played an important role in MM progression. Moreover, ectopic HERC4 expression decreased MM proliferation in vitro, and delayed xenograft tumor growth in vivo. Therefore, modulation of c-Maf ubiquitination by targeting HERC4 may represent a new therapeutic modality for MM.
Collapse
|