1
|
Tang F, Cui Q. Diverse roles of aldolase enzymes in cancer development, drug resistance and therapeutic approaches as moonlighting enzymes. Med Oncol 2024; 41:224. [PMID: 39120781 DOI: 10.1007/s12032-024-02470-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Aldolase enzymes, particularly ALDOA, ALDOB, and ALDOC, play a crucial role in the development and progression of cancer. While the aldolase family is mainly known for its involvement in the glycolysis pathway, these enzymes also have various pathological and physiological functions through distinct signaling pathways such as Wnt/β-catenin, EGFR/MAPK, Akt, and HIF-1α. This has garnered increased attention in recent years and shed light on other sides of this enzyme. Potential therapeutic strategies targeting aldolases include using siRNA, inhibitors like naphthol AS-E phosphate and TX-2098, and natural compounds such as HDPS-4II and L-carnosine. Additionally, anticancer peptides derived from ALDOA, like P04, can potentially increase cancer cells' sensitivity to chemotherapy. Aldolases also affect cancer drug resistance by different approaches, making them good therapeutic targets. In this review, we extensively explore the role of aldolase enzymes in various types of cancers in proliferation, invasion, migration, and drug resistance; we also significantly explore the possible treatment considering aldolase function.
Collapse
Affiliation(s)
- Fan Tang
- General Surgery Department, Xinhua Hospital of Yili Kazak Autonomous Prefecture, YiLi, 835000, China
| | - Qingyang Cui
- Department of Interventional Oncology, Xinhua Hospital of Yili Kazak Autonomous Prefecture, YiLi, 835000, China.
| |
Collapse
|
2
|
Liu J, Han D, Xuan J, Xie J, Wang W, Zhou Q, Chen K. COP9 signalosome complex is a prognostic biomarker and corresponds with immune infiltration in hepatocellular carcinoma. Aging (Albany NY) 2024; 16:5264-5287. [PMID: 38466642 PMCID: PMC11006475 DOI: 10.18632/aging.205646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024]
Abstract
Hepatocellular carcinoma (HCC) is among the most common deadly tumors but still lacks specific biomarkers for diagnosis, prognosis, and treatment guidance. The COP9 signalosome (COPS) is an essential regulator of the ubiquitin conjugation pathway upregulated in various cancers. We evaluated the contributions of COPS subunits to HCC tumorigenesis and their utility for prognosis. We comprehensively evaluated the tumor expression pattern and tumorigenic functions of COPS subunits using The Cancer Genome Atlas (TCGA), The Human Protein Atlas and immunohistochemistry. Kaplan-Meier, Cox regression, ROC curve, and nomogram analyses were used to assess the predictive values of COPS subunits for clinical outcome. Expression levels of COPS subunits were significantly upregulated in HCC tissues, which predicted shorter overall survival (OS). Further, Cox regression analysis identified COPS5, COPS7B, and COPS9 as independent prognostic biomarkers for OS. High mutation rates were also found in COPS subunits. Functional network analysis indicated that COPS and neighboring genes regulate 'protein neddylation', 'protein deneddylation', and 'protein ubiquitination'. The COPS PPI included strong interactions with p53, CUL1/2/3/4, and JUN. Moreover, the correlations between COPS subunit expression levels and tumor immune cell infiltration rates were examined using TIMER, TISIDB, ssGSEA, and ESTIMATE packages. COPS subunits expression levels were positively correlated with specific tumor immune cell infiltration rates, immunoregulator expression levels, and microsatellite instability in HCC. Finally, knockout of COPS6 and COPS9 in HCC cells reduced while overexpression enhanced proliferation rate and metastasis capacity. Our study revealed that COPS potential biomarker for unfavorable HCC prognosis and indicators of immune infiltration, tumorigenicity, and metastasis.
Collapse
Affiliation(s)
- Jiahui Liu
- Department of Clinical Laboratory, Zhongshan City People’s Hospital, The Affiliated Zhongshan Hospital of Sun Yat-Sen University, Zhongshan 528400, Guangdong, China
- Laboratory of Basic Medical Science, General Hospital of Southern Theater Command of PLA, Guangzhou 510000, Guangdong, China
| | - Dexing Han
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong, China
| | - Junfeng Xuan
- Laboratory of Basic Medical Science, General Hospital of Southern Theater Command of PLA, Guangzhou 510000, Guangdong, China
| | - Jinye Xie
- Department of Clinical Laboratory, Zhongshan City People’s Hospital, The Affiliated Zhongshan Hospital of Sun Yat-Sen University, Zhongshan 528400, Guangdong, China
| | - Weijia Wang
- Department of Clinical Laboratory, Zhongshan City People’s Hospital, The Affiliated Zhongshan Hospital of Sun Yat-Sen University, Zhongshan 528400, Guangdong, China
| | - Quan Zhou
- Laboratory of Basic Medical Science, General Hospital of Southern Theater Command of PLA, Guangzhou 510000, Guangdong, China
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong, China
| | - Kang Chen
- Department of Clinical Laboratory, Zhongshan City People’s Hospital, The Affiliated Zhongshan Hospital of Sun Yat-Sen University, Zhongshan 528400, Guangdong, China
| |
Collapse
|
3
|
Du WQ, Zhu ZM, Jiang X, Kang MJ, Pei DS. COPS6 promotes tumor progression and reduces CD8 + T cell infiltration by repressing IL-6 production to facilitate tumor immune evasion in breast cancer. Acta Pharmacol Sin 2023; 44:1890-1905. [PMID: 37095198 PMCID: PMC10462724 DOI: 10.1038/s41401-023-01085-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/28/2023] [Indexed: 04/26/2023]
Abstract
Due to poor T cell infiltration, tumors evade immune surveillance. Increased CD8+ T cell infiltration in breast cancer suggests a satisfactory response to immunotherapy. COPS6 has been identified as an oncogene, but its role in regulating antitumor immune responses has not been defined. In this study, we investigated the impact of COPS6 on tumor immune evasion in vivo. Tumor transplantation models were established in C57BL/6 J mice and BALB/c nude mice. Flow cytometry was conducted to identify the role of COPS6 on tumor-infiltrating CD8+ T cells. By analyzing the TCGA and GTEx cohort, we found that COPS6 expression was significantly up-regulated in a variety of cancers. In human osteosarcoma cell line U2OS and non-small cell lung cancer cell line H1299, we showed that p53 negatively regulated COPS6 promoter activity. In human breast cancer MCF-7 cells, COPS6 overexpression stimulated p-AKT expression as well as the proliferation and malignant transformation of tumor cells, whereas knockdown of COPS6 caused opposite effects. Knockdown of COPS6 also significantly suppressed the growth of mouse mammary cancer EMT6 xenografts in BALB/c nude mice. Bioinformatics analysis suggested that COPS6 was a mediator of IL-6 production in the tumor microenvironment and a negative regulator of CD8+ T cell tumor infiltration in breast cancer. In C57BL6 mice bearing EMT6 xenografts, COPS6 knockdown in the EMT6 cells increased the number of tumor-infiltrating CD8+ T cells, while knockdown of IL-6 in COPS6KD EMT6 cells diminished tumor infiltrating CD8+ T cells. We conclude that COPS6 promotes breast cancer progression by reducing CD8+ T cell infiltration and function via the regulation of IL-6 secretion. This study clarifies the role of p53/COPS6/IL-6/CD8+ tumor infiltrating lymphocytes signaling in breast cancer progression and immune evasion, opening a new path for development of COPS6-targeting therapies to enhance tumor immunogenicity and treat immunologically "cold" breast cancer.
Collapse
Affiliation(s)
- Wen-Qi Du
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhi-Man Zhu
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Xin Jiang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Meng-Jie Kang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Dong-Sheng Pei
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
4
|
Lu Y, Zhang Y, Wang X, Zhang H, Zhu Y, Zhang J, Sha H, Zou R, Gan Y, Sui Y, Wang J, Du T, Wu J, Feng J. Aldolase A Promotes Colorectal Cancer Progression through Targeting COPS6 and Regulating MAPK Signaling Pathway. DISEASE MARKERS 2023; 2023:1702125. [PMID: 37457886 PMCID: PMC10344634 DOI: 10.1155/2023/1702125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/26/2022] [Accepted: 01/04/2023] [Indexed: 07/18/2023]
Abstract
Colorectal cancer (CRC) is a serious threat to human health, and its underlying mechanisms remain to be further explored. Aldolase A (ALDOA) has received increasing attention for its reported association with multiple cancers, but the role and mechanisms of ALDOA in CRC are still unclear. In the current study, high expression levels and enzymatic activity of ALDOA were detected in CRC tissues and cell lines, indicating the clinical significance of ALDOA in human CRC. In addition, silencing ALDOA significantly impaired the proliferation and metastasis of CRC cells in vitro and in vivo. Mechanistically, immunoprecipitation assays and mass spectrometry analysis identified the binding protein COPS6 of ALDOA. Furthermore, the promoting effects of upregulated ALDOA on CRC cell proliferation and metastasis were inhibited by COPS6 depletion, demonstrating COPS6 was required for ALDOA in mediating CRC progress. Moreover, the epithelial-mesenchymal transition (EMT) program and MAPK signaling pathway were found to be activated by ALDOA overexpression as well. In summary, our findings suggested that ALDOA facilitated the proliferation and metastasis of CRC by binding and regulating COPS6, inducing EMT, and activating the mitogen-activated protein kinase (MAPK) signaling pathway. The present study provided evidence for ALDOA as a promising potential biomarker for CRC.
Collapse
Affiliation(s)
- Ya Lu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan Zhang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyue Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Zhang
- Department of Endoscopy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), China
| | - Yue Zhu
- Nanjing Jinling Hospital, China
| | - Junying Zhang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Huanhuan Sha
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Renrui Zou
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yujie Gan
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Sui
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Tongde Du
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jianzhong Wu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jifeng Feng
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Beydogan AB, Coskun Yazici ZM, Bolkent S. Influences of calorie restriction and lipopolysaccharide therapy on inflammation, cytokine response, and cell proliferation in pancreatic adenocarcinoma mouse model. J Biochem Mol Toxicol 2023; 37:e23250. [PMID: 36281497 DOI: 10.1002/jbt.23250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/05/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
The study aimed to investigate the effects of lipopolysaccharide (LPS) alone and in combination with calorie restriction (CR) on the pancreatic tissues in C57BL/6 mice modeled with pancreatic ductal adenocarcinoma (PDAC). Forty male C57BL/6 mice (10-13 weeks old) were divided into five groups; LPS, LPS + CR, PDAC, PDAC + LPS, and PDAC + LPS + CR. Nuclear factor kappa B (NF-κβ), interleukin-6 (IL-6), and c-Jun N-terminal kinases (JNK) mRNA expression levels were measured in pancreatic tissues. NF-κβ, IL-6, JNK, and proliferating cell nuclear antigen (PCNA) peptide levels were determined by immunohistochemistry. Oxidative stress markers and antioxidant enzyme activities were determined spectrophotometrically. TH1/TH2 cytokine measurements were determined by a flow cytometer. It was detected that the number of PCNA immune + cells in the PDAC + LPS + CR group was significantly lower than in the PDAC and PDAC + LPS groups (p < 0.01, p < 0.05 respectively). PDAC + LPS + CR group's plasma interferon-gamma (IFN-γ), IL-6, IL-2, tumor necrosis factor-alpha, IL-3, and IL-4 levels were found to be significantly lower than the PDAC group (p < 0.01, p < 0.001, p < 0.01, p < 0.05, p < 0.01, and p < 0.05 respectively). According to our findings, the combination of low-dose LPS and 40% CR was found to be more effective in PDAC model mice.
Collapse
Affiliation(s)
- Alisa B Beydogan
- Department of Medical Biology, Faculty of Cerrahpasa Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeynep M Coskun Yazici
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul, Turkey
| | - Sema Bolkent
- Department of Medical Biology, Faculty of Cerrahpasa Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
6
|
Du W, Zhang R, Muhammad B, Pei D. Targeting the COP9 signalosome for cancer therapy. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0605. [PMID: 35315259 PMCID: PMC9196064 DOI: 10.20892/j.issn.2095-3941.2021.0605] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/18/2022] [Indexed: 11/24/2022] Open
Abstract
The COP9 signalosome (CSN) is a highly conserved protein complex composed of 8 subunits (CSN1 to CSN8). The individual subunits of the CSN play essential roles in cell proliferation, tumorigenesis, cell cycle regulation, DNA damage repair, angiogenesis, and microenvironmental homeostasis. The CSN complex has an intrinsic metalloprotease that removes the ubiquitin-like activator NEDD8 from cullin-RING ligases (CRLs). Binding of neddylated CRLs to CSN is sensed by CSN4 and communicated to CSN5 with the assistance of CSN6, thus leading to the activation of deneddylase. Therefore, CSN is a crucial regulator at the intersection between neddylation and ubiquitination in cancer progression. Here, we summarize current understanding of the roles of individual CSN subunits in cancer progression. Furthermore, we explain how the CSN affects tumorigenesis through regulating transcription factors and the cell cycle. Finally, we discuss individual CSN subunits as potential therapeutic targets to provide new directions and strategies for cancer therapy.
Collapse
Affiliation(s)
- Wenqi Du
- Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, China
| | - Ruicheng Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - Bilal Muhammad
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - Dongsheng Pei
- Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
7
|
Qi B, Liu H, Zhou Q, Ji L, Shi X, Wei Y, Gu Y, Mizushima A, Xia S. An immune-related lncRNA signature for the prognosis of pancreatic adenocarcinoma. Aging (Albany NY) 2021; 13:18806-18826. [PMID: 34285140 PMCID: PMC8351726 DOI: 10.18632/aging.203323] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022]
Abstract
Recent evidence suggests that aberrant expression of long non-coding RNA (lncRNA) can drive the initiation and progression of malignancies. However, little is known about the prognostic potential of lncRNA. We aimed at constructing a lncRNA-based signature to improve the prognosis prediction of pancreatic adenocarcinoma (PAAD). The PAAD samples with clinical information were obtained from The Cancer Genome Atlas and International Cancer Genome Consortium. We established an eight-IRlncRNA signature in a training cohort. The prognostic value of eight-IRlncRNA signature was validated in two distinct cohorts when compared to other four prognostic models. We continued to analyze its independence in subgroups by univariate and multivariate Cox regression. We constructed a nomogram for clinicopathologic features and 1-, 3-, and 5-year overall survival performance. Moreover, Gene set enrichment analysis and Gene Set Variation Analysis distinguished the typical functions between high- and low-risk groups. In addition, we further observed the different correlations of immune cell between eight IRlncRNAs. Eight-IRlncRNA signature appears to be a good performer to predict the survival capability of PAAD patients, and the nomogram will enable PAAD patients to be more accurately managed in clinical practice.
Collapse
Affiliation(s)
- Bing Qi
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Han Liu
- College of Stomatology, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Qi Zhou
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Li Ji
- Department of Gastroenterology, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning, China
| | - Xueying Shi
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Yushan Wei
- Department of Scientific Research, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Yajun Gu
- School of Medical Laboratory, Tianjin Medical University, Tianjin 300000, Tianjin, China
| | - Akio Mizushima
- Department of Palliative Medicine, Graduate School of Medicine, Juntendo University, Tokyo 1138421, Japan
| | - Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| |
Collapse
|
8
|
STING agonist and IDO inhibitor combination therapy inhibits tumor progression in murine models of colorectal cancer. Cell Immunol 2021; 366:104384. [PMID: 34182334 DOI: 10.1016/j.cellimm.2021.104384] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 05/05/2021] [Accepted: 05/19/2021] [Indexed: 11/22/2022]
Abstract
Despite impressive clinical success, cancer immunotherapy based on immune checkpoint blockade remains ineffective in colorectal cancer (CRC). Stimulator of interferon genes (STING) is a novel potential target and STING agonists have shown potential anti-tumor efficacy. Combined therapy based on synergistic mechanism can overcome the resistance. However, STING agonists-based combination therapies are deficient. We designed different immunotherapy combinations, including STING agonist, indoleamine 2,3 dioxygenase (IDO) inhibitor and PD-1 blockade, with purpose of exploring which option can effectively inhibit CRC growth. To further explore the possible reasons of therapeutic effectiveness, we observed the combination therapy in C57BL/6Tmem173gt mice. Our findings demonstrated that STING agonist diABZI combined with IDO inhibitor 1-MT significantly inhibited tumor growth, even better than the three-drug combination, promoted the recruitment of CD8+ T cells and dendritic cells, and decreased the infiltration of myeloid-derived suppressor cells. We conclude that diABZI combined with 1-MT is a promising option for CRC.
Collapse
|
9
|
Zhang Y, Hou J, Shi S, Du J, Liu Y, Huang P, Li Q, Liu L, Hu H, Ji Y, Guo L, Shi Y, Liu Y, Cui H. CSN6 promotes melanoma proliferation and metastasis by controlling the UBR5-mediated ubiquitination and degradation of CDK9. Cell Death Dis 2021; 12:118. [PMID: 33483464 PMCID: PMC7822921 DOI: 10.1038/s41419-021-03398-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/17/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022]
Abstract
As a critical subunit of the constitutive photomorphogenesis 9 (COP9) signalosome (CSN), CSN6 is upregulated in some human cancers and plays critical roles in tumorigenesis and progression, but its biological functions and molecular mechanisms in melanoma remain unknown. Our study showed that CSN6 expression was upregulated in melanoma patients and cells, and correlated with poor survival in melanoma patients. In melanoma cells, CSN6 knockdown remarkably inhibited cell proliferation, tumorigenicity, migration, and invasion, whereas CSN6 recovery rescued the proliferative and metastatic abilities. Notably, we identified that CSN6 stabilized CDK9 expression by reducing CDK9 ubiquitination levels, thereby activating CDK9-mediated signaling pathways. In addition, our study described a novel CSN6-interacting E3 ligase UBR5, which was negatively regulated by CSN6 and could regulate the ubiquitination and degradation of CDK9 in melanoma cells. Furthermore, in CSN6-knockdown melanoma cells, UBR5 knockdown abrogated the effects caused by CSN6 silencing, suggesting that CSN6 activates the UBR5/CDK9 pathway to promote melanoma cell proliferation and metastasis. Thus, this study illustrates the mechanism by which the CSN6-UBR5-CDK9 axis promotes melanoma development, and demonstrate that CSN6 may be a potential biomarker and anticancer target in melanoma.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China
| | - Jianbing Hou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China.,Cancer center, Medical Research Institute, Southwest University, 400716, Chongqing, China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, 400716, Chongqing, China
| | - Shaomin Shi
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China
| | - Juan Du
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China
| | - Yudong Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China.,Cancer center, Medical Research Institute, Southwest University, 400716, Chongqing, China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, 400716, Chongqing, China
| | - Pan Huang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China.,Cancer center, Medical Research Institute, Southwest University, 400716, Chongqing, China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, 400716, Chongqing, China
| | - Qian Li
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China
| | - Lichao Liu
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China
| | - Huanrong Hu
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China
| | - Yacong Ji
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China
| | - Leiyang Guo
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China
| | - Yaqiong Shi
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China
| | - Yaling Liu
- Department of Dermatology, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400715, Chongqing, China. .,Cancer center, Medical Research Institute, Southwest University, 400716, Chongqing, China. .,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China. .,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, 400716, Chongqing, China.
| |
Collapse
|
10
|
Fu H, Zhang Y, Chen Y, Chen J, Chen P. CSN1 facilitates proliferation and migration of hepatocellular carcinoma cells by upregulating cyclin A2 expression. Mol Med Rep 2021; 23:46. [PMID: 33200803 PMCID: PMC7705997 DOI: 10.3892/mmr.2020.11684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022] Open
Abstract
Constitutive photomorphogenesis 9 signalosome subunit 1 (CSN1) plays an important role in the ubiquitin-proteasome pathway and regulates various cellular processes, such as the cell cycle and DNA repair. The CSN complex consists of eight subunits (CSN1 to CSN8) and regulates the tumorigenesis of a variety of tumor types. However, the exact role of CSN1 in hepatocellular carcinoma (HCC) remains unclear. The present study evaluated the expression and biological effects of CSN1 in HCC tissue samples and cell lines. CSN1 was significantly overexpressed in HCC tissue and cell lines, compared with their normal counterparts. In patients with HCC, elevated CSN1 levels correlated with tumor size, tumor metastasis and tumor stage. Loss‑of‑function assays indicated that CSN1 knockdown inhibited the proliferation and migration HCC cells. In addition, CSN1 promoted the expression of cyclin A2 in a ubiquitination‑independent manner. Lastly, xenograft experiments indicated that CSN1 promoted HCC tumor growth in vivo. The present study suggested that CSN1 inhibition could represent a potential approach for the prevention of HCC progression and metastasis.
Collapse
Affiliation(s)
- Hangwei Fu
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Yida Zhang
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Yin Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
- Department of Gynecology and Obstetrics, The 958th Hospital, Southwest Hospital, Army Medical University, Chongqing 400020, P.R. China
| | - Junying Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Ping Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| |
Collapse
|