1
|
Peng Y, Qi X, Ding L, Huang J, Liu Y, Zheng R, Fu Y, Yin L, Deng T, Ye Y, Chen S, Li X. SKP2 inhibition activates tumor cell-intrinsic immunity by inducing DNA replication stress and genomic instability. Br J Cancer 2025; 132:81-92. [PMID: 39582087 PMCID: PMC11723935 DOI: 10.1038/s41416-024-02909-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND S-phase kinase-associated protein 2 (SKP2) is a typical oncogene aberrantly overexpressing in a variety of cancer types, but it remains elusive whether SKP2 regulates the antitumor immunity of triple-negative breast cancer. METHODS The efficacy of anti-PD-1 was evaluated in the orthotopic xenografts of immunocompetent mice models. The infiltration of cytotoxic T cells in tumor microenvironment(TME) were assessed by immunofluorescence staining. The levels of pro-inflammatory chemokines were analyzed by ELISA. The protein interaction was analyzed by co-immunoprecipitation and GST pull-down. The genomic instability was analyzed by fluorescent microscopy. RESULTS SKP2 inhibition significantly improved the antitumor efficacy of immune checkpoint blockade (ICB). Furthermore, SKP2 inhibition activated the cGAS/STING signal pathway and induced the secretion of pro-inflammatory chemokines, thereby promoting cytotoxic T cell infiltration. Additionally, we identified CDC6, a DNA replication licensing factor as a novel substrate of SKP2 in addition to CDT1. SKP2 induced protein degradation of CDC6 and CDT1 through the ubiquitin-proteasome pathway. Conversely, SKP2 inhibition elevated CDC6 and CDT1 protein levels, which caused DNA aberrant replication, DNA damage and genomic instability, thereby resulting in the accumulation of cytosolic DNA, activating cGAS/STING signaling pathway and improving antitumor immunity. CONCLUSION SKP2 may be used as an effective therapeutic target to enable ICB antitumor immunotherapy. SOCIAL MEDIA Peng et al. found that SKP2 inhibition improved the antitumor immunotherapy by activating tumor cell-intrinsic immunity, thereby providing evidences that SKP2 may be used as an effective therapeutic target to enable ICB antitumor immunotherapy.
Collapse
Affiliation(s)
- Yuchong Peng
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
- Institute of Integrative Neurology of integrated traditional Chinese and Western Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
| | - Xuli Qi
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Liuyang Ding
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Jingjing Huang
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Youhong Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Rirong Zheng
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Yongming Fu
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
- Institute of Integrative Neurology of integrated traditional Chinese and Western Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
| | - Linglong Yin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Tanggang Deng
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Yubing Ye
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Size Chen
- Department of Tumor Immunity, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
| | - Xiong Li
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China.
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China.
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China.
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
2
|
Pan C, Pan C, Chen L, Lin A. Machine learning analysis of oxidative stress-related phenotypes for specific gene screening in ovarian cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:4763-4775. [PMID: 39162393 DOI: 10.1002/tox.24321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Oxidative stress serves a crucial role in tumor development. However, the relationship between ovarian cancer and oxidative stress remains unknown. We aimed to create an oxidative stress-related prognostic signature to enhance the prognosis prediction of CC patients using bioinformatics. METHODS The genes differentially expressed and associated with oxidative stress were extracted with the help of "limma" packages. The model for prognosis was created using Multivariate Cox regression analysis to determine the risk related to the genes related to oxidative stress. Patients were categorized as low-risk or high-risk based on the median score. The receiver operation characteristic (ROC) and survival curves were used to evaluate the predictive effect of the prognostic signature. We utilized quantitative real-time PCR to assess the expression levels of key genes associated with oxidative stress in ovarian cancer cell lines (SKOV3, OVCAR3, and HeyA8) and normal ovarian epithelial cells (HOSEpiC). RESULTS A signature comprising seven genes associated with oxidative stress was developed to prognosticate patients with ovarian cancer. Overall survival (OS) of the patient having CC was determined using Kaplan-Meier analysis. It was found that patient with a higher risk score had lower OS than the low-risk score. The signature of genes associated with oxidative stress was found to be independently prognostic for 1, 2, and 3 years. Further research found that the expression levels of nine hub genes had a strong association with patient outcomes. Our analysis revealed a higher expression of CX3CR1 in ovarian cancer cell lines compared with normal cells. CONCLUSIONS To deploy a novel oxidative stress-related prognostic signature as an independent biomarker in cervical cancer, we developed and validated it.
Collapse
Affiliation(s)
- Chenxiang Pan
- Department of Women's Oncology, Shuangyu Campus, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Chunyu Pan
- Department of Women's Oncology, Shuangyu Campus, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Lili Chen
- Department of Women's Oncology, Shuangyu Campus, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Aidi Lin
- Department of Women's Oncology, Shuangyu Campus, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| |
Collapse
|
3
|
Xu W, Zhang Y, Su Y, Li L, Yang X, Wang L, Gao H. USP9X regulates the proliferation, survival, migration and invasion of gastric cancer cells by stabilizing MTH1. BMC Gastroenterol 2024; 24:239. [PMID: 39075342 PMCID: PMC11288101 DOI: 10.1186/s12876-024-03321-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND MutT homolog 1 (MTH1) sanitizes oxidized dNTP pools to promote the survival of cancer cells and its expression is frequently upregulated in cancers. Polyubiquitination stabilizes MTH1 to facilitate the proliferation of melanoma cells, suggesting the ubiquitin system controls the stability and function of MTH1. However, whether ubiquitination regulates MTH1 in gastric cancers has not been well defined. This study aims to investigate the interaction between MTH1 and a deubiquitinase, USP9X, in regulating the proliferation, survival, migration, and invasion of gastric cancer cells. METHODS The interaction between USP9X and MTH1 was evaluated by co-immunoprecipitation (co-IP) in HGC-27 gastric cancer cells. siRNAs were used to interfere with USP9X expression in gastric cancer cell lines HGC-27 and MKN-45. MTT assays were carried out to examine the proliferation, propidium iodide (PI) and 7-AAD staining assays were performed to assess the cell cycle, Annexin V/PI staining assays were conducted to examine the apoptosis, and transwell assays were used to determine the migration and invasion of control, USP9X-deficient, and USP9X-deficient plus MTH1-overexpressing HGC-27 and MKN-45 gastric cancer cells. RESULTS Co-IP data show that USP9X interacts with and deubiquitinates MTH1. Overexpression of USP9X elevates MTH1 protein level by downregulating its ubiquitination, while knockdown of USP9X has the opposite effect on MTH1. USP9X deficiency in HGC-27 and MKN-45 cells causes decreased proliferation, cell cycle arrest, extra apoptosis, and defective migration and invasion, which could be rescued by excessive MTH1. CONCLUSION USP9X interacts with and stabilizes MTH1 to promote the proliferation, survival, migration and invasion of gastric cancer cells.
Collapse
Affiliation(s)
- Wenji Xu
- Digestive System Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Yaping Zhang
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, No. 34, Zhongshan North Road, Licheng District, Quanzhou, 362000, China
| | - Yingrui Su
- Nuclear Medicine Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Libin Li
- Digestive System Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xinxia Yang
- Digestive System Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Lixing Wang
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, No. 34, Zhongshan North Road, Licheng District, Quanzhou, 362000, China.
| | - Hongzhi Gao
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, No. 34, Zhongshan North Road, Licheng District, Quanzhou, 362000, China.
- Neurosurgery Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| |
Collapse
|
4
|
Jiang Y, Ni S, Xiao B, Jia L. Function, mechanism and drug discovery of ubiquitin and ubiquitin-like modification with multiomics profiling for cancer therapy. Acta Pharm Sin B 2023; 13:4341-4372. [PMID: 37969742 PMCID: PMC10638515 DOI: 10.1016/j.apsb.2023.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/21/2023] [Accepted: 07/17/2023] [Indexed: 11/17/2023] Open
Abstract
Ubiquitin (Ub) and ubiquitin-like (Ubl) pathways are critical post-translational modifications that determine whether functional proteins are degraded or activated/inactivated. To date, >600 associated enzymes have been reported that comprise a hierarchical task network (e.g., E1-E2-E3 cascade enzymatic reaction and deubiquitination) to modulate substrates, including enormous oncoproteins and tumor-suppressive proteins. Several strategies, such as classical biochemical approaches, multiomics, and clinical sample analysis, were combined to elucidate the functional relations between these enzymes and tumors. In this regard, the fundamental advances and follow-on drug discoveries have been crucial in providing vital information concerning contemporary translational efforts to tailor individualized treatment by targeting Ub and Ubl pathways. Correspondingly, emphasizing the current progress of Ub-related pathways as therapeutic targets in cancer is deemed essential. In the present review, we summarize and discuss the functions, clinical significance, and regulatory mechanisms of Ub and Ubl pathways in tumorigenesis as well as the current progress of small-molecular drug discovery. In particular, multiomics analyses were integrated to delineate the complexity of Ub and Ubl modifications for cancer therapy. The present review will provide a focused and up-to-date overview for the researchers to pursue further studies regarding the Ub and Ubl pathways targeted anticancer strategies.
Collapse
Affiliation(s)
| | | | - Biying Xiao
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
5
|
Liu L, Hou Q, Chen B, Lai X, Wang H, Liu H, Wu L, Liu S, Luo K, Liu J. Identification of molecular subgroups and establishment of risk model based on the response to oxidative stress to predict overall survival of patients with lung adenocarcinoma. Eur J Med Res 2023; 28:333. [PMID: 37689745 PMCID: PMC10492289 DOI: 10.1186/s40001-023-01290-5] [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: 07/05/2023] [Accepted: 08/14/2023] [Indexed: 09/11/2023] Open
Abstract
OBJECTIVE Oxidative stress is associated with the occurrence and development of lung cancer. However, the specific association between lung cancer and oxidative stress is unclear. This study aimed to investigate the role of oxidative stress in the progression and prognosis of lung adenocarcinoma (LUAD). METHODS The gene expression profiles and corresponding clinical information were collected from GEO and TCGA databases. Differentially expressed oxidative stress-related genes (OSRGs) were identified between normal and tumor samples. Consensus clustering was applied to identify oxidative stress-related molecular subgroups. Functional enrichment analysis, GSEA, and GSVA were performed to investigate the potential mechanisms. xCell was used to assess the immune status of the subgroups. A risk model was developed by the LASSO algorithm and validated using TCGA-LUAD, GSE13213, and GSE30219 datasets. RESULTS A total of 40 differentially expressed OSRGs and two oxidative stress-associated subgroups were identified. Enrichment analysis revealed that cell cycle-, inflammation- and oxidative stress-related pathways varied significantly in the two subgroups. Furthermore, a risk model was developed and validated based on the OSRGs, and findings indicated that the risk model exhibits good prediction and diagnosis values for LUAD patients. CONCLUSION The risk model based on the oxidative stress could act as an effective prognostic tool for LUAD patients. Our findings provided novel genetic biomarkers for prognosis prediction and personalized clinical treatment for LUAD patients.
Collapse
Affiliation(s)
- Linzhuang Liu
- Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, 518036, Guangdong, China
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Qinghua Hou
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Baorong Chen
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Xiyi Lai
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Hanwen Wang
- Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, 518036, Guangdong, China
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Haozhen Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Liusheng Wu
- Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, 518036, Guangdong, China
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Sheng Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Kelin Luo
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
6
|
Zhou H, Zhou L, Guan Q, Hou X, Wang C, Liu L, Wang J, Yu X, Li W, Liu H. Skp2-mediated MLKL degradation confers cisplatin-resistant in non-small cell lung cancer cells. Commun Biol 2023; 6:805. [PMID: 37532777 PMCID: PMC10397346 DOI: 10.1038/s42003-023-05166-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most prevalent type of cancer and the leading cause of cancer-related death. Chemotherapeutic resistance is a major obstacle in treating NSCLC patients. Here, we discovered that the E3 ligase Skp2 is overexpressed, accompanied by the downregulation of necroptosis-related regulator MLKL in human NSCLC tissues and cell lines. Knockdown of Skp2 inhibited viability, anchorage-independent growth, and in vivo tumor development of NSCLC cells. We also found that the Skp2 protein is negatively correlated with MLKL in NSCLC tissues. Moreover, Skp2 is increased and accompanied by an upregulation of MLKL ubiquitination and degradation in cisplatin-resistant NSCLC cells. Accordingly, inhibition of Skp2 partially restores MLKL and sensitizes NSCLC cells to cisplatin in vitro and in vivo. Mechanistically, Skp2 interacts and promotes ubiquitination-mediated degradation of MLKL in cisplatin-resistant NSCLC cells. Our results provide evidence of an Skp2-dependent mechanism regulating MLKL degradation and cisplatin resistance, suggesting that targeting Skp2-ubiquitinated MLKL degradation may overcome NSCLC chemoresistance.
Collapse
Affiliation(s)
- Huiling Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, The Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qing Guan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xuyang Hou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Cong Wang
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lijun Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jian Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xinfang Yu
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Haidan Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Becker AL, Indra AK. Oxidative Stress in Melanoma: Beneficial Antioxidant and Pro-Oxidant Therapeutic Strategies. Cancers (Basel) 2023; 15:cancers15113038. [PMID: 37297001 DOI: 10.3390/cancers15113038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Cutaneous melanoma ranks as the fifth most common cancer in the United States and represents one of the deadliest forms of skin cancer. While recent advances in systemic targeted therapies and immunotherapies have positively impacted melanoma survival, the survival rate of stage IV melanoma remains at a meager 32%. Unfortunately, tumor resistance can impede the effectiveness of these treatments. Oxidative stress is a pivotal player in all stages of melanoma progression, with a somewhat paradoxical function that promotes tumor initiation but hinders vertical growth and metastasis in later disease. As melanoma progresses, it employs adaptive mechanisms to lessen oxidative stress in the tumor environment. Redox metabolic rewiring has been implicated in acquired resistance to BRAF/MEK inhibitors. A promising approach to enhance the response to therapy involves boosting intracellular ROS production using active biomolecules or targeting enzymes that regulate oxidative stress. The complex interplay between oxidative stress, redox homeostasis, and melanomagenesis can also be leveraged in a preventive context. The purpose of this review is to provide an overview of oxidative stress in melanoma, and how the antioxidant system may be manipulated in a therapeutic context for improved efficacy and survival.
Collapse
Affiliation(s)
- Alyssa L Becker
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR 97331, USA
- John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI 96813, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR 97331, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- Department of Biochemistry and Biophysics, Oregon State University (OSU), Corvallis, OR 97331, USA
- Linus Pauling Science Center, Oregon State University (OSU), Corvallis, OR 97331, USA
- Department of Dermatology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| |
Collapse
|
8
|
Li Q, Zhang W. Progress in Anticancer Drug Development Targeting Ubiquitination-Related Factors. Int J Mol Sci 2022; 23:ijms232315104. [PMID: 36499442 PMCID: PMC9737479 DOI: 10.3390/ijms232315104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 12/05/2022] Open
Abstract
Ubiquitination is extensively involved in critical signaling pathways through monitoring protein stability, subcellular localization, and activity. Dysregulation of this process results in severe diseases including malignant cancers. To develop drugs targeting ubiquitination-related factors is a hotspot in research to realize better therapy of human diseases. Ubiquitination comprises three successive reactions mediated by Ub-activating enzyme E1, Ub-conjugating enzyme E2, and Ub ligase E3. As expected, multiple ubiquitination enzymes have been highlighted as targets for anticancer drug development due to their dominant effect on tumorigenesis and cancer progression. In this review, we discuss recent progresses in anticancer drug development targeting enzymatic machinery components.
Collapse
|
9
|
Xu M, Liu Y, Wan HL, Wong AM, Ding X, You W, Lo WS, Ng KKC, Wong N. Overexpression of nucleotide metabolic enzyme DUT in hepatocellular carcinoma potentiates a therapeutic opportunity through targeting its dUTPase activity. Cancer Lett 2022; 548:215898. [PMID: 36075487 DOI: 10.1016/j.canlet.2022.215898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 07/24/2022] [Accepted: 08/24/2022] [Indexed: 11/26/2022]
Abstract
Uracil misincorporation during DNA replication is a major cell toxic event, of which cancer cells overcome by activating the dUTPase enzyme. The DUT gene is the only known dUTPase in human. Despite reports on common upregulations in cancers, the role of DUT in human hepatocellular carcinoma (HCC) remains largely undetermined. In this study, we investigated the mechanism underlying DUT biology in HCC and tumor susceptibility to drug targeting dUTPase. Overexpression of DUT was found in 42% of HCC tumors and correlated with advanced stage HCC. Knockout of DUT in HCC cell lines showed suppressed proliferation through cell cycle arrest and a spontaneous induction of DNA damage. A protective effect from oxidative stress was also demonstrated in both knockout and overexpression DUT assays. Transcriptome analysis highlighted the NF-κB survival signaling as the downstream effector pathway of DUT in overriding oxidative stress-induced cell death. Interestingly, stably expressed DUT in liver progenitor organoids conferred drug resistance to TKI Sorafenib. Targeting dUTPase activity by TAS-114, could potentiate suppression of HCC growth that synergized with Sorafenib for better treatment sensitivity. In conclusion, upregulated DUT represents a nucleotide metabolic weakness and therapeutic opportunity in HCC.
Collapse
Affiliation(s)
- Mingjing Xu
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yue Liu
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ho Lee Wan
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Alissa M Wong
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Xiaofan Ding
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wenxing You
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wing Sze Lo
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Kelvin K-C Ng
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Nathalie Wong
- Department of Surgery, Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
10
|
Role of K63-linked ubiquitination in cancer. Cell Death Dis 2022; 8:410. [PMID: 36202787 PMCID: PMC9537175 DOI: 10.1038/s41420-022-01204-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/16/2022] [Accepted: 09/26/2022] [Indexed: 11/08/2022]
Abstract
Ubiquitination is a critical type of post-translational modifications, of which K63-linked ubiquitination regulates interaction, translocation, and activation of proteins. In recent years, emerging evidence suggest involvement of K63-linked ubiquitination in multiple signaling pathways and various human diseases including cancer. Increasing number of studies indicated that K63-linked ubiquitination controls initiation, development, invasion, metastasis, and therapy of diverse cancers. Here, we summarized molecular mechanisms of K63-linked ubiquitination dictating different biological activities of tumor and highlighted novel opportunities for future therapy targeting certain regulation of K63-linked ubiquitination in tumor.
Collapse
|
11
|
Hsu CC, Peng D, Cai Z, Lin HK. AMPK signaling and its targeting in cancer progression and treatment. Semin Cancer Biol 2022; 85:52-68. [PMID: 33862221 PMCID: PMC9768867 DOI: 10.1016/j.semcancer.2021.04.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022]
Abstract
The intrinsic mechanisms sensing the imbalance of energy in cells are pivotal for cell survival under various environmental insults. AMP-activated protein kinase (AMPK) serves as a central guardian maintaining energy homeostasis by orchestrating diverse cellular processes, such as lipogenesis, glycolysis, TCA cycle, cell cycle progression and mitochondrial dynamics. Given that AMPK plays an essential role in the maintenance of energy balance and metabolism, managing AMPK activation is considered as a promising strategy for the treatment of metabolic disorders such as type 2 diabetes and obesity. Since AMPK has been attributed to aberrant activation of metabolic pathways, mitochondrial dynamics and functions, and epigenetic regulation, which are hallmarks of cancer, targeting AMPK may open up a new avenue for cancer therapies. Although AMPK is previously thought to be involved in tumor suppression, several recent studies have unraveled its tumor promoting activity. The double-edged sword characteristics for AMPK as a tumor suppressor or an oncogene are determined by distinct cellular contexts. In this review, we will summarize recent progress in dissecting the upstream regulators and downstream effectors for AMPK, discuss the distinct roles of AMPK in cancer regulation and finally offer potential strategies with AMPK targeting in cancer therapy.
Collapse
Affiliation(s)
- Che-Chia Hsu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA
| | - Danni Peng
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA
| | - Zhen Cai
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA.
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
12
|
Zhang M, Du G, Li Z, Li D, Li W, Li H, Gao X, Tang Z. An Oxidative Stress-Related Genes Signature for Predicting Survival in Bladder Cancer: Based on TCGA Database and Bioinformatics. Int J Gen Med 2022; 15:2645-2667. [PMID: 35300137 PMCID: PMC8922338 DOI: 10.2147/ijgm.s348945] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/23/2022] [Indexed: 12/24/2022] Open
Abstract
Background Oxidative stress (OS) responses have been linked to oncogenesis and tumor progression and have recently been regarded as a potential strategy for tumor therapy. However, OS-related therapeutic targets have not been identified to date in the bladder cancer (BC). Methods The mRNA expression and clinical data of BC were downloaded from the public database. Prognostic risk score signature was constructed using LASSO Cox regression analysis. External validation was performed in GSE15307 cohort. ESTIMATE, CIBERSORT, and ssGSEA algorithm were used to analyze immune cell infiltration and immune microenvironment. Next, functional enrichment analysis was performed to elucidate the mechanism underlying the signature. Additionally, we performed a nomogram to forecast the survival rate of individual BC patients. Results An OS-related genes (OSRGs) signature was constructed. Overall survival was lower in the high-risk group than in the low-risk group, according to survival analyses. The area under the curve (AUC) of ROC curves further validated the prognostic signature’s strong prediction performance in these two cohorts. The risk score was verified as an independent risk factor for BC by independent prognostic analysis. Moreover, as compared to TNM stage alone, a nomogram that integrated the risk score with TNM stage showed a much superior predictive value. Immune infiltration and tumor microenvironment studies indicated that immune cells and functions may play a significant role in carcinogenesis and development. The levels of expression of prognostic genes were shown to be substantially linked with drug sensitivity. Conclusion We developed a novel OSRGs signature for predicting overall survival and impacting the immune status in patients with BC. New nomogram can help clinicians predict the survival rate of BC patients. These findings shed new light on the potential usage of OSRGs signature in BC patients.
Collapse
Affiliation(s)
- Min Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Gang Du
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Zhengtian Li
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Dehui Li
- Department of Burns and Plastic, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Weichao Li
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Hening Li
- Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Xingxin Gao
- Department of Burns and Plastic, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Zhanhong Tang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People’s Republic of China
- Correspondence: Zhanhong Tang; Xingxin Gao, Email ;
| |
Collapse
|
13
|
Kuttikrishnan S, Prabhu KS, Khan AQ, Alali FQ, Ahmad A, Uddin S. Thiostrepton inhibits growth and induces apoptosis by targeting FoxM1/SKP2/MTH1 axis in B-precursor acute lymphoblastic leukemia cells. Leuk Lymphoma 2021; 62:3170-3180. [PMID: 34369229 DOI: 10.1080/10428194.2021.1957873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Forkhead box M1 (FoxM1) is a transcription factor that plays an important role in the etiology of many cancers, however, its role has not been elucidated in B-precursor acute lymphoblastic leukemia (B-pre-ALL). In the current study, we showed that the downregulation of FoxM1 by its inhibitor thiostrepton inhibited cell viability and induced caspase-dependent apoptosis in a panel of B-pre-ALL cell lines. Thiostrepton led downregulation of FoxM1 accompanied by decreased expression of Aurora kinase A, B, matrix metalloproteinases, and oncogene SKP2 as well as MTH1. Downregulation of the FoxM1/SKP2/MTH1 axis led to increase in the Bax/Bcl2 ratio and suppression of antiapoptotic proteins. Thiostrepton-mediated apoptosis was prevented by N-acetyl cysteine, a scavenger of reactive oxygen species. Co-treatment of B-pre-ALL with subtoxic doses of thiostrepton and bortezomib potentiated the proapoptotic action. Altogether, our results suggest that targeting FoxM1expression could be an attractive strategy for the treatment of B-pre-ALL.
Collapse
Affiliation(s)
- Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,College of Pharmacy, Qatar University, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Feras Q Alali
- College of Pharmacy, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
14
|
Lv S, Liu M, Xu L, Zhang C. Downregulation of decidual SKP2 is associated with human recurrent miscarriage. Reprod Biol Endocrinol 2021; 19:88. [PMID: 34116705 PMCID: PMC8194034 DOI: 10.1186/s12958-021-00775-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 06/01/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Recurrent miscarriage (RM) is a very frustrating problem for both couples and clinicians. To date, the etiology of RM remains poorly understood. Decidualization plays a critical role in implantation and the maintenance of pregnancy, and its deficiency is closely correlated with RM. The F-box protein S-phase kinase associated protein 2 (SKP2) is a key component of the SCF-type E3 ubiquitin ligase complex, which is critically involved in ErbB family-induced Akt ubiquitination, aerobic glycolysis and tumorigenesis. SKP2 is pivotal for reproduction, and SKP2-deficient mice show impaired ovarian development and reduced fertility. METHODS Here, we investigated the expression and function of SKP2 in human decidualization and its relation with RM. A total of 40 decidual samples were collected. Quantitative PCR analysis, western blot analysis and immunohistochemistry analysis were performed to analyze the differential expression of SKP2 between RM and control cells. For in vitro induction of decidualization, both HESCs (human endometrial stromal cells) cell line and primary ESCs (endometrial stromal cells) were used to analyze the effects of SKP2 on decidualization via siRNA transfection. RESULTS Compared to normal pregnant women, the expression of SKP2 was reduced in the decidual tissues from individuals with RM. After in vitro induction of decidualization, knockdown of SKP2 apparently attenuated the decidualization of HESCs and resulted in the downregulation of HOXA10 and FOXM1, which are essential for normal human decidualization. Moreover, our experiments demonstrated that SKP2 silencing reduced the expression of its downstream target GLUT1. CONCLUSIONS Our study indicates a functional role of SKP2 in RM: downregulation of SKP2 in RM leads to impaired decidualization and downregulation of GLUT1 and consequently predisposes individuals to RM.
Collapse
Affiliation(s)
- Shijian Lv
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Mei Liu
- Department of Obstetrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42 Wenhua Xi Road, Jinan, 250011, Shandong, China
| | - Lizhen Xu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
| |
Collapse
|
15
|
Yang Y, Long X, Li K, Li G, Yu X, Wen P, Luo J, Tian X, Zhao J. Development and validation of an oxidative stress-associated prognostic risk model for melanoma. PeerJ 2021; 9:e11258. [PMID: 33976978 PMCID: PMC8063880 DOI: 10.7717/peerj.11258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/22/2021] [Indexed: 11/20/2022] Open
Abstract
Background Oxidative stress (OS) is key to various diseases and is implicated in cancer progression and oncogenesis. However, the potential diagnostic value of OS-related genes in skin cutaneous melanoma (SKCM) remains unclear. Methods We used data of RNA sequencing from 471 tumor tissues and one healthy tissue acquired from The Cancer Genome Atlas (TCGA)-SKCM cohort. The Genome Tissue Expression database was used to acquire transcriptome data from 812 healthy samples. OS-related genes that were differentially expressed between SKCM and healthy samples were investigated and 16 prognosis-associated OS genes were identified. The prognostic risk model was built using univariate and Cox multivariate regressions. The prognostic value of the hub genes was validated in the GSE65904 cohort, which included 214 SKCM patients. Results The overall survival rate of SKCM patients in the high-risk group was decreased compared to the low-risk group. In both TCGA and GSE65904 cohorts, the ROC curves suggested that our prognostic risk model was more accurate than other clinicopathological characteristics to diagnose SKCM. Moreover, risk score and nomograms associated with the expression of hub genes were developed. These presented reiterated our prognostic risk model. Altogether, this study provides novel insights with regards to the pathogenesis of SKCM. The 16 hub genes identified may help in SKCM prognosis and individualized clinical treatment.
Collapse
Affiliation(s)
- Yu Yang
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xuan Long
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Kun Li
- The Second People's Hospital of Guiyang, Guiyang, China
| | - Guiyun Li
- The Second People's Hospital of Guiyang, Guiyang, China
| | - Xiaohong Yu
- The Second People's Hospital of Guiyang, Guiyang, China
| | - Ping Wen
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jun Luo
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | | | - Jinmin Zhao
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
16
|
Vitry G, Paulin R, Grobs Y, Lampron MC, Shimauchi T, Lemay SE, Tremblay E, Habbout K, Awada C, Bourgeois A, Nadeau V, Paradis R, Breuils-Bonnet S, Roux-Dalvai F, Orcholski M, Potus F, Provencher S, Boucherat O, Bonnet S. Oxidized DNA Precursors Cleanup by NUDT1 Contributes to Vascular Remodeling in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2021; 203:614-627. [PMID: 33021405 DOI: 10.1164/rccm.202003-0627oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Pulmonary arterial hypertension (PAH) is a life-threatening condition characterized by abnormally elevated pulmonary pressures and right ventricular failure. Excessive proliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs) is one of the most important drivers of vascular remodeling in PAH, for which available treatments have limited effectiveness.Objectives: To gain insights into the mechanisms leading to the development of the disease and identify new actionable targets.Methods: Protein expression profiling was conducted by two-dimensional liquid chromatography coupled to tandem mass spectrometry in isolated PASMCs from controls and patients with PAH. Multiple molecular, biochemical, and pharmacologic approaches were used to decipher the role of NUDT1 (nudrix hyrolase 1) in PAH.Measurements and Main Results: Increased expression of the detoxifying DNA enzyme NUDT1 was detected in cells and tissues from patients with PAH and animal models. In vitro, molecular or pharmacological inhibition of NUDT1 in PAH-PASMCs induced accumulation of oxidized nucleotides in the DNA, irresolvable DNA damage (comet assay), disruption of cellular bioenergetics (Seahorse), and cell death (terminal deoxynucleotidyl transferase dUTP nick end labeling assay). In two animal models with established PAH (i.e., monocrotaline and Sugen/hypoxia-treated rats), pharmacological inhibition of NUDT1 using (S)-Crizotinib significantly decreased pulmonary vascular remodeling and improved hemodynamics and cardiac function.Conclusions: Our results indicate that, by overexpressing NUDT1, PAH-PASMCs hijack persistent oxidative stress in preventing incorporation of oxidized nucleotides into DNA, thus allowing the cell to escape apoptosis and proliferate. Given that NUDT1 inhibitors are under clinical investigation for cancer, they may represent a new therapeutic option for PAH.
Collapse
Affiliation(s)
- Géraldine Vitry
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Roxane Paulin
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and.,Department of Medicine and
| | - Yann Grobs
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Marie-Claude Lampron
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Sarah-Eve Lemay
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Eve Tremblay
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Karima Habbout
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Charifa Awada
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Alice Bourgeois
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Valérie Nadeau
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Renée Paradis
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | | | - Mark Orcholski
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - François Potus
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and.,Department of Medicine and
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and.,Department of Medicine and
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, Québec City, Quebec, Canada; and.,Department of Medicine and
| |
Collapse
|
17
|
Wu X, Zhao J. Novel oxidative stress-related prognostic biomarkers for melanoma associated with tumor metastasis. Medicine (Baltimore) 2021; 100:e24866. [PMID: 33663112 PMCID: PMC7909214 DOI: 10.1097/md.0000000000024866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/30/2021] [Indexed: 01/05/2023] Open
Abstract
Skin cutaneous melanoma (SKCM) is a prevalent skin cancer whose metastatic form is dangerous due to its high morbidity and mortality. Previous studies have systematically established the vital role of oxidative stress (OS) in melanoma progression. This study aimed to identify prognostic OS genes closely associated with SKCM and illustrate their potential mechanisms. Transcriptome data and corresponding clinical traits of patients with SKCM were retrieved from The Cancer Genome Atlas and Gene Expression Omnibus databases. A weighted gene co-expression network analysis was conducted to identify relationships between clinical features and OS genes in specific modules. Subsequently, Cox regression analysis was performed on candidate OS genes; four hub prognosis-associated OS genes (AKAP9, VPS13C, ACSL4, and HMOX2) were identified to construct a prognostic model. After a series of bioinformatics analysis, our prognostic model was identified significantly associated with the overall survival of patients with SKCM and metastatic ability of the cancer. Furthermore, our risk model demonstrated improved diagnostic accuracy in the Cancer Genome Atlas and Gene Expression Omnibus cohorts. In addition, we established 2 nomograms based on either risk score or hub genes, which displayed favorable discriminating ability for SKCM. Our results provide novel insight into the potential applications of OS-associated genes in SKCM.
Collapse
Affiliation(s)
- Xianpei Wu
- Department of Orthopedics Trauma and Hand Surgery
| | - Jinmin Zhao
- Department of Orthopedics Trauma and Hand Surgery
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration
- Guangxi Collaborative Innovation Center for Biomedicine
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| |
Collapse
|
18
|
Wright RHG, Beato M. Role of the NUDT Enzymes in Breast Cancer. Int J Mol Sci 2021; 22:2267. [PMID: 33668737 PMCID: PMC7956304 DOI: 10.3390/ijms22052267] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Despite global research efforts, breast cancer remains the leading cause of cancer death in women worldwide. The majority of these deaths are due to metastasis occurring years after the initial treatment of the primary tumor and occurs at a higher frequency in hormone receptor-positive (Estrogen and Progesterone; HR+) breast cancers. We have previously described the role of NUDT5 (Nudix-linked to moiety X-5) in HR+ breast cancer progression, specifically with regards to the growth of breast cancer stem cells (BCSCs). BCSCs are known to be the initiators of epithelial-to-mesenchyme transition (EMT), metastatic colonization, and growth. Therefore, a greater understanding of the proteins and signaling pathways involved in the metastatic process may open the door for therapeutic opportunities. In this review, we discuss the role of NUDT5 and other members of the NUDT family of enzymes in breast and other cancer types. We highlight the use of global omics data based on our recent phosphoproteomic analysis of progestin signaling pathways in breast cancer cells and how this experimental approach provides insight into novel crosstalk mechanisms for stratification and drug discovery projects aiming to treat patients with aggressive cancer.
Collapse
Affiliation(s)
- Roni H. G. Wright
- Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08003 Barcelona, Spain
| | - Miguel Beato
- Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain
- Department of Life Science, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| |
Collapse
|
19
|
Integrated analysis identifies oxidative stress genes associated with progression and prognosis in gastric cancer. Sci Rep 2021; 11:3292. [PMID: 33558567 PMCID: PMC7870842 DOI: 10.1038/s41598-021-82976-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/27/2021] [Indexed: 12/25/2022] Open
Abstract
Oxidative stress (OS) reactions are reported to be associated with oncogenesis and tumor progression. However, little is known about the potential diagnostic value of OS in gastric cancer (GC). This study identified hub OS genes associated with the prognosis and progression of GC and illustrated the underlying mechanisms. The transcriptome data and corresponding GC clinical information were collected from The Cancer Genome Atlas (TCGA) database. Aberrantly expressed OS genes between tumors and adjacent normal tissues were screened, and 11 prognosis-associated genes were identified with a series of bioinformatic analyses and used to construct a prognostic model. These genes were validated in the Gene Expression Omnibus (GEO) database. Furthermore, weighted gene co-expression network analysis (WGCNA) was subsequently conducted to identify the most significant hub genes for the prediction of GC progression. Analysis revealed that a good prognostic model was constructed with a better diagnostic accuracy than other clinicopathological characteristics in both TCGA and GEO cohorts. The model was also significantly associated with the overall survival of patients with GC. Meanwhile, a nomogram based on the risk score was established, which displayed a favorable discriminating ability for GC. In the WGCNA analysis, 13 progression-associated hub OS genes were identified that were also significantly associated with the progression of GC. Furthermore, functional and gene ontology (GO) analyses were performed to reveal potential pathways enriched with these genes. These results provide novel insights into the potential applications of OS-associated genes in patients with GC.
Collapse
|
20
|
Qiu X, Hou QH, Shi QY, Jiang HX, Qin SY. Identification of Hub Prognosis-Associated Oxidative Stress Genes in Pancreatic Cancer Using Integrated Bioinformatics Analysis. Front Genet 2020; 11:595361. [PMID: 33363572 PMCID: PMC7753072 DOI: 10.3389/fgene.2020.595361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background Intratumoral oxidative stress (OS) has been associated with the progression of various tumors. However, OS has not been considered a candidate therapeutic target for pancreatic cancer (PC) owing to the lack of validated biomarkers. Methods We compared gene expression profiles of PC samples and the transcriptome data of normal pancreas tissues from The Cancer Genome Atlas (TCGA) and Genome Tissue Expression (GTEx) databases to identify differentially expressed OS genes in PC. PC patients’ gene profile from the Gene Expression Omnibus (GEO) database was used as a validation cohort. Results A total of 148 differentially expressed OS-related genes in PC were used to construct a protein-protein interaction network. Univariate Cox regression analysis, least absolute shrinkage, selection operator analysis revealed seven hub prognosis-associated OS genes that served to construct a prognostic risk model. Based on integrated bioinformatics analyses, our prognostic model, whose diagnostic accuracy was validated in both cohorts, reliably predicted the overall survival of patients with PC and cancer progression. Further analysis revealed significant associations between seven hub gene expression levels and patient outcomes, which were validated at the protein level using the Human Protein Atlas database. A nomogram based on the expression of these seven hub genes exhibited prognostic value in PC. Conclusion Our study provides novel insights into PC pathogenesis and provides new genetic markers for prognosis prediction and clinical treatment personalization for PC patients.
Collapse
Affiliation(s)
- Xin Qiu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qin-Han Hou
- Department of Neurosurgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Qiu-Yue Shi
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hai-Xing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shan-Yu Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
21
|
Li DN, Yang CC, Li J, Ou Yang QG, Zeng LT, Fan GQ, Liu TH, Tian XY, Wang JJ, Zhang H, Dai DP, Cui J, Cai JP. The high expression of MTH1 and NUDT5 promotes tumor metastasis and indicates a poor prognosis in patients with non-small-cell lung cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118895. [PMID: 33096144 DOI: 10.1016/j.bbamcr.2020.118895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/03/2020] [Accepted: 10/15/2020] [Indexed: 12/25/2022]
Abstract
MutT Homolog 1 (MTH1) is a mammalian 8-oxodGTPase for sanitizing oxidative damage to the nucleotide pool. Nudix type 5 (NUDT5) also sanitizes 8-oxodGDP in the nucleotide pool. The role of MTH1 and NUDT5 in non-small-cell lung cancer (NSCLC) progression and metastasis remains unclear. In the present study, we reported that MTH1 and NUDT5 were upregulated in NSCLC cell lines and tissues, and higher levels of MTH1 or NUDT5 were associated with tumor metastasis and a poor prognosis in patients with NSCLC. Their suppression also restrained tumor growth and lung metastasis in vivo and significantly inhibited NSCLC cell migration, invasion, cell proliferation and cell cycle progression while promoting apoptosis in vitro. The opposite effects were observed in vitro following MTH1 or NUDT5 rescue. In addition, the upregulation of MTH1 or NUDT5 enhanced the MAPK pathway and PI3K/AKT activity. Furthermore, MTH1 and NUDT5 induce epithelial-mesenchymal transition both in vitro and in vivo. These results highlight the essential role of MTH1 and NUDT5 in NSCLC tumor tumorigenesis and metastasis as well as their functions as valuable markers of the NSCLC prognosis and potential therapeutic targets.
Collapse
Affiliation(s)
- Dan-Ni Li
- Peking University Fifth School of Clinical Medicine, Beijing Hospital, Beijing, PR China
| | - Cheng-Cheng Yang
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Jin Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO.1 DaHua Road, Dong Dan, Beijing 100730, PR China
| | - Qiu-Geng Ou Yang
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Lv-Tao Zeng
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO.1 DaHua Road, Dong Dan, Beijing 100730, PR China
| | - Guo-Qing Fan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO.1 DaHua Road, Dong Dan, Beijing 100730, PR China
| | - Teng-Hui Liu
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xin-Yuan Tian
- Peking University Fifth School of Clinical Medicine, Beijing Hospital, Beijing, PR China
| | - Jing-Jing Wang
- Peking University Fifth School of Clinical Medicine, Beijing Hospital, Beijing, PR China
| | - He Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO.1 DaHua Road, Dong Dan, Beijing 100730, PR China
| | - Da-Peng Dai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO.1 DaHua Road, Dong Dan, Beijing 100730, PR China
| | - Ju Cui
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO.1 DaHua Road, Dong Dan, Beijing 100730, PR China
| | - Jian-Ping Cai
- Peking University Fifth School of Clinical Medicine, Beijing Hospital, Beijing, PR China; The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, NO.1 DaHua Road, Dong Dan, Beijing 100730, PR China.
| |
Collapse
|
22
|
Feng YC, Liu XY, Teng L, Ji Q, Wu Y, Li JM, Gao W, Zhang YY, La T, Tabatabaee H, Yan XG, Jamaluddin MFB, Zhang D, Guo ST, Scott RJ, Liu T, Thorne RF, Zhang XD, Jin L. c-Myc inactivation of p53 through the pan-cancer lncRNA MILIP drives cancer pathogenesis. Nat Commun 2020; 11:4980. [PMID: 33020477 PMCID: PMC7536215 DOI: 10.1038/s41467-020-18735-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
The functions of the proto-oncoprotein c-Myc and the tumor suppressor p53 in controlling cell survival and proliferation are inextricably linked as “Yin and Yang” partners in normal cells to maintain tissue homeostasis: c-Myc induces the expression of ARF tumor suppressor (p14ARF in human and p19ARF in mouse) that binds to and inhibits mouse double minute 2 homolog (MDM2) leading to p53 activation, whereas p53 suppresses c-Myc through a combination of mechanisms involving transcriptional inactivation and microRNA-mediated repression. Nonetheless, the regulatory interactions between c-Myc and p53 are not retained by cancer cells as is evident from the often-imbalanced expression of c-Myc over wildtype p53. Although p53 repression in cancer cells is frequently associated with the loss of ARF, we disclose here an alternate mechanism whereby c-Myc inactivates p53 through the actions of the c-Myc-Inducible Long noncoding RNA Inactivating P53 (MILIP). MILIP functions to promote p53 polyubiquitination and turnover by reducing p53 SUMOylation through suppressing tripartite-motif family-like 2 (TRIML2). MILIP upregulation is observed amongst diverse cancer types and is shown to support cell survival, division and tumourigenicity. Thus our results uncover an inhibitory axis targeting p53 through a pan-cancer expressed RNA accomplice that links c-Myc to suppression of p53. c-Myc and p53 operate in a negative feedback manner to maintain cellular homeostasis. Here, the authors report a long noncoding RNA, MILIP as a downstream target of c-Myc and that MILIP represses p53 to support tumorigenicity.
Collapse
Affiliation(s)
- Yu Chen Feng
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Xiao Ying Liu
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Liu Teng
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Qiang Ji
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Yongyan Wu
- Department of Otolaryngology, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, the first affiliated hospital, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jin Ming Li
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Wei Gao
- Department of Otolaryngology, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, the first affiliated hospital, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yuan Yuan Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Ting La
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Hessam Tabatabaee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Xu Guang Yan
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - M Fairuz B Jamaluddin
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Didi Zhang
- Department of Orthopaedics, John Hunter Hospital, Hunter New England Health, Newcastle, 2305, NSW, Australia
| | - Su Tang Guo
- Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Taiyuan, 030013, Shanxi, China
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, 2750, NSW, Australia
| | - Rick F Thorne
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia.,Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia. .,Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China.
| | - Lei Jin
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China. .,School of Medicine and Public Health, The University of Newcastle, Newcastle, 2308, NSW, Australia.
| |
Collapse
|
23
|
Das I, Gad H, Bräutigam L, Pudelko L, Tuominen R, Höiom V, Almlöf I, Rajagopal V, Hansson J, Helleday T, Egyházi Brage S, Warpman Berglund U. AXL and CAV-1 play a role for MTH1 inhibitor TH1579 sensitivity in cutaneous malignant melanoma. Cell Death Differ 2020; 27:2081-2098. [PMID: 31919461 PMCID: PMC7308409 DOI: 10.1038/s41418-019-0488-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023] Open
Abstract
Cutaneous malignant melanoma (CMM) is the deadliest form of skin cancer and clinically challenging due to its propensity to develop therapy resistance. Reactive oxygen species (ROS) can induce DNA damage and play a significant role in CMM. MTH1 protein protects from ROS damage and is often overexpressed in different cancer types including CMM. Herein, we report that MTH1 inhibitor TH1579 induced ROS levels, increased DNA damage responses, caused mitotic arrest and suppressed CMM proliferation leading to cell death both in vitro and in an in vivo xenograft CMM zebrafish disease model. TH1579 was more potent in abrogating cell proliferation and inducing cell death in a heterogeneous co-culture setting when compared with CMM standard treatments, vemurafenib or trametinib, showing its broad anticancer activity. Silencing MTH1 alone exhibited similar cytotoxic effects with concomitant induction of mitotic arrest and ROS induction culminating in cell death in most CMM cell lines tested, further emphasizing the importance of MTH1 in CMM cells. Furthermore, overexpression of receptor tyrosine kinase AXL, previously demonstrated to contribute to BRAF inhibitor resistance, sensitized BRAF mutant and BRAF/NRAS wildtype CMM cells to TH1579. AXL overexpression culminated in increased ROS levels in CMM cells. Moreover, silencing of a protein that has shown opposing effects on cell proliferation, CAV-1, decreased sensitivity to TH1579 in a BRAF inhibitor resistant cell line. AXL-MTH1 and CAV-1-MTH1 mRNA expressions were correlated as seen in CMM clinical samples. Finally, TH1579 in combination with BRAF inhibitor exhibited a more potent cell killing effect in BRAF mutant cells both in vitro and in vivo. In summary, we show that TH1579-mediated efficacy is independent of BRAF/NRAS mutational status but dependent on the expression of AXL and CAV-1.
Collapse
Affiliation(s)
- Ishani Das
- Department of Oncology-Pathology, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Helge Gad
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, S-171 64, Stockholm, Sweden
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, S10 2RX, UK
| | - Lars Bräutigam
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Linda Pudelko
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Rainer Tuominen
- Department of Oncology-Pathology, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Ingrid Almlöf
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Varshni Rajagopal
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, S-171 64, Stockholm, Sweden
- Department of Oncology, Karolinska University Hospital, S-171 76, Stockholm, Sweden
| | - Thomas Helleday
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, S-171 64, Stockholm, Sweden
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, S10 2RX, UK
| | - Suzanne Egyházi Brage
- Department of Oncology-Pathology, Karolinska Institutet, S-171 64, Stockholm, Sweden
| | - Ulrika Warpman Berglund
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, S-171 64, Stockholm, Sweden.
| |
Collapse
|
24
|
Yumimoto K, Yamauchi Y, Nakayama KI. F-Box Proteins and Cancer. Cancers (Basel) 2020; 12:cancers12051249. [PMID: 32429232 PMCID: PMC7281081 DOI: 10.3390/cancers12051249] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022] Open
Abstract
Controlled protein degradation is essential for the operation of a variety of cellular processes including cell division, growth, and differentiation. Identification of the relations between ubiquitin ligases and their substrates is key to understanding the molecular basis of cancer development and to the discovery of novel targets for cancer therapeutics. F-box proteins function as the substrate recognition subunits of S-phase kinase-associated protein 1 (SKP1)−Cullin1 (CUL1)−F-box protein (SCF) ubiquitin ligase complexes. Here, we summarize the roles of specific F-box proteins that have been shown to function as tumor promoters or suppressors. We also highlight proto-oncoproteins that are targeted for ubiquitylation by multiple F-box proteins, and discuss how these F-box proteins are deployed to regulate their cognate substrates in various situations.
Collapse
|
25
|
Avitan-Hersh E, Feng Y, Oknin Vaisman A, Abu Ahmad Y, Zohar Y, Zhang T, Lee JS, Lazar I, Sheikh Khalil S, Feiler Y, Kluger H, Kahana C, Brown K, Ruppin E, Ronai ZA, Orian A. Regulation of eIF2α by RNF4 Promotes Melanoma Tumorigenesis and Therapy Resistance. J Invest Dermatol 2020; 140:2466-2477. [PMID: 32360601 DOI: 10.1016/j.jid.2020.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/24/2020] [Accepted: 04/06/2020] [Indexed: 01/07/2023]
Abstract
Among the hallmarks of melanoma are impaired proteostasis and rapid development of resistance to targeted therapy that represent a major clinical challenge. However, the molecular machinery that links these processes is unknown. Here we describe that by stabilizing key melanoma oncoproteins, the ubiquitin ligase RNF4 promotes tumorigenesis and confers resistance to targeted therapy in melanoma cells, xenograft mouse models, and patient samples. In patients, RNF4 protein and mRNA levels correlate with poor prognosis and with resistance to MAPK inhibitors. Remarkably, RNF4 tumorigenic properties, including therapy resistance, require the translation initiation factor initiation elongation factor alpha (eIF2α). RNF4 binds, ubiquitinates, and stabilizes the phosphorylated eIF2α (p-eIF2α) but not activating transcription factor 4 or C/EBP homologous protein that mediates the eIF2α-dependent integrated stress response. In accordance, p-eIF2α levels were significantly elevated in high-RNF4 patient-derived melanomas. Thus, RNF4 and p-eIF2α establish a positive feed-forward loop connecting oncogenic translation and ubiquitin-dependent protein stabilization in melanoma.
Collapse
Affiliation(s)
- Emily Avitan-Hersh
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center, Technion-Israel Institute of Technology, Haifa, Israel; Rambam Health Care Campus, Haifa, Israel
| | - Yongmei Feng
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Avital Oknin Vaisman
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yamen Abu Ahmad
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yaniv Zohar
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center, Technion-Israel Institute of Technology, Haifa, Israel; Rambam Health Care Campus, Haifa, Israel
| | - Tongwu Zhang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Joo Sang Lee
- Cancer Data Science Lab, National Cancer Institute, NIH, Maryland, USA; Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Ikrame Lazar
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Saeed Sheikh Khalil
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yulia Feiler
- Deprtament of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| | - Harriet Kluger
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chaim Kahana
- Deprtament of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| | - Kevin Brown
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, NIH, Maryland, USA
| | - Ze'ev A Ronai
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center, Technion-Israel Institute of Technology, Haifa, Israel; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Amir Orian
- Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
26
|
La T, Jin L, Liu XY, Song ZH, Farrelly M, Feng YC, Yan XG, Zhang YY, Thorne RF, Zhang XD, Teng L. Cylindromatosis Is Required for Survival of a Subset of Melanoma Cells. Oncol Res 2020; 28:385-398. [PMID: 32252875 PMCID: PMC7851542 DOI: 10.3727/096504020x15861709922491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The deubiquitinase cylindromatosis (CYLD) functions as a tumor suppressor inhibiting cell proliferation in many cancer types including melanoma. Here we present evidence that a proportion of melanoma cells are nonetheless addicted to CYLD for survival. The expression levels of CYLD varied widely in melanoma cell lines and melanomas in vivo, with a subset of melanoma cell lines and melanomas displaying even higher levels of CYLD than melanocyte lines and nevi, respectively. Strikingly, although short hairpin RNA (shRNA) knockdown of CYLD promoted, as anticipated, cell proliferation in some melanoma cell lines, it reduced cell viability in a fraction of melanoma cell lines with relatively high levels of CYLD expression and did not impinge on survival and proliferation in a third type of melanoma cell lines. The decrease in cell viability caused by CYLD knockdown was due to induction of apoptosis, as it was associated with activation of the caspase cascade and was abolished by treatment with a general caspase inhibitor. Mechanistic investigations demonstrated that induction of apoptosis by CYLD knockdown was caused by upregulation of receptor-interacting protein kinase 1 (RIPK1) that was associated with elevated K63-linked polyubiquitination of the protein, indicating that CYLD is critical for controlling RIPK1 expression in these cells. Of note, microRNA (miR) profiling showed that miR-99b-3p that was predicted to target the 3′-untranslated region (3′-UTR) of the CYLD mRNA was reduced in melanoma cell lines with high levels of CYLD compared with melanocyte lines. Further functional studies confirmed that the reduction in miR-99b-3p expression was responsible for the increased expression of CYLD in a highly cell line-specific manner. Taken together, these results reveal an unexpected role of CYLD in promoting survival of a subset of melanoma cells and uncover the heterogeneity of CYLD expression and its biological significance in melanoma.
Collapse
Affiliation(s)
- Ting La
- Translational Research Institute, Henan Provincial Peoples Hospital, Academy of Medical Science, Zhengzhou UniversityZhengzhouP.R. China
| | - Lei Jin
- Translational Research Institute, Henan Provincial Peoples Hospital, Academy of Medical Science, Zhengzhou UniversityZhengzhouP.R. China
| | - Xiao Ying Liu
- Translational Research Institute, Henan Provincial Peoples Hospital, Academy of Medical Science, Zhengzhou UniversityZhengzhouP.R. China
| | - Ze Hua Song
- Translational Research Institute, Henan Provincial Peoples Hospital, Academy of Medical Science, Zhengzhou UniversityZhengzhouP.R. China
| | - Margaret Farrelly
- School of Biomedical Sciences and Pharmacy, The University of NewcastleCallaghan, NSWAustralia
| | - Yu Chen Feng
- School of Biomedical Sciences and Pharmacy, The University of NewcastleCallaghan, NSWAustralia
| | - Xu Guang Yan
- School of Biomedical Sciences and Pharmacy, The University of NewcastleCallaghan, NSWAustralia
| | - Yuan Yuan Zhang
- School of Biomedical Sciences and Pharmacy, The University of NewcastleCallaghan, NSWAustralia
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial Peoples Hospital, Academy of Medical Science, Zhengzhou UniversityZhengzhouP.R. China
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial Peoples Hospital, Academy of Medical Science, Zhengzhou UniversityZhengzhouP.R. China
| | - Liu Teng
- Translational Research Institute, Henan Provincial Peoples Hospital, Academy of Medical Science, Zhengzhou UniversityZhengzhouP.R. China
| |
Collapse
|
27
|
Yumimoto K, Nakayama KI. Recent insight into the role of FBXW7 as a tumor suppressor. Semin Cancer Biol 2020; 67:1-15. [PMID: 32113998 DOI: 10.1016/j.semcancer.2020.02.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/15/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022]
Abstract
FBXW7 (also known as Fbw7, Sel10, hCDC4, or hAgo) is a tumor suppressor and the most frequently mutated member of the F-box protein family in human cancers. FBXW7 functions as the substrate recognition component of an SCF-type E3 ubiquitin ligase. It specifically controls the proteasome-mediated degradation of many oncoproteins such as c-MYC, NOTCH, KLF5, cyclin E, c-JUN, and MCL1. In this review, we summarize the molecular and biological features of FBXW7 and its substrates as well as the impact of mutations of FBXW7 on cancer development. We also address the clinical potential of anticancer therapy targeting FBXW7.
Collapse
Affiliation(s)
- Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan.
| |
Collapse
|
28
|
Cai Z, Moten A, Peng D, Hsu CC, Pan BS, Manne R, Li HY, Lin HK. The Skp2 Pathway: A Critical Target for Cancer Therapy. Semin Cancer Biol 2020; 67:16-33. [PMID: 32014608 DOI: 10.1016/j.semcancer.2020.01.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/22/2020] [Accepted: 01/25/2020] [Indexed: 12/16/2022]
Abstract
Strictly regulated protein degradation by ubiquitin-proteasome system (UPS) is essential for various cellular processes whose dysregulation is linked to serious diseases including cancer. Skp2, a well characterized component of Skp2-SCF E3 ligase complex, is able to conjugate both K48-linked ubiquitin chains and K63-linked ubiquitin chains on its diverse substrates, inducing proteasome mediated proteolysis or modulating the function of tagged substrates respectively. Overexpression of Skp2 is observed in various human cancers associated with poor survival and adverse therapeutic outcomes, which in turn suggests that Skp2 engages in tumorigenic activity. To that end, the oncogenic properties of Skp2 are demonstrated by various genetic mouse models, highlighting the potential of Skp2 as a target for tackling cancer. In this article, we will describe the downstream substrates of Skp2 as well as upstream regulators for Skp2-SCF complex activity. We will further summarize the comprehensive oncogenic functions of Skp2 while describing diverse strategies and therapeutic platforms currently available for developing Skp2 inhibitors.
Collapse
Affiliation(s)
- Zhen Cai
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA.
| | - Asad Moten
- National Capital Consortium, Department of Defense, Washington DC, 20307, USA; Institute for Complex Systems, HealthNovations International, Houston, TX, 77089, USA; Center for Cancer Research, National Institutes of Health, Bethesda, MD, 20814, USA; Center on Genomics, Vulnerable Populations, and Health Disparities, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Danni Peng
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Che-Chia Hsu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Rajeshkumar Manne
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Hong-Yu Li
- University of Arkansas for Medical Sciences, College of Pharmacy, Division of Pharmaceutical Science, 200 South Cedar, Little Rock AR 72202, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA; Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
29
|
Reactive Oxygen Species Are Involved in the Development of Gastric Cancer and Gastric Cancer-Related Depression through ABL1-Mediated Inflammation Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5813985. [PMID: 31396300 PMCID: PMC6664690 DOI: 10.1155/2019/5813985] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 05/30/2019] [Indexed: 12/19/2022]
Abstract
Background The mechanisms of crosstalk between depression and gastric cancer (GC) remain ill defined. Given that reactive oxygen species (ROS) is involved in the pathophysiology of both GC and depression, we try to explore the activities of ROS in the development of GC and GC-related depression. Methods 110 patients with newly diagnosed GC were recruited in our study. The clinical characteristics of these patients were recorded. Inflammation and oxidative stress markers were detected by ELISA. The depression status of patients with GC was assessed during follow-up. The association between ROS, ABL1, and inflammation factors was evaluated in H2O2-treated GC cell lines and The Cancer Genome Atlas (TCGA) database. The effect of ABL1 on inflammation was detected with Imatinib/Nilotinib-treated GC cell lines. A chronic mild stress- (CMS-) induced patient-derived xenograft (PDX) mice model was established to assess the crosstalk between depression and GC. Results Depression was correlated with poor prognosis of patients with GC. GC patients with depression were under a high level of oxidative status as well as dysregulated inflammation. In the CMS-induced GC PDX mice model, CMS could facilitate the development of GC. Additionally, tumor bearing could induce depressive-like behaviors of mice. With the treatment of ROS, the activities of ABL1 and inflammatory signaling were enhanced both in vitro and in vivo, and blocking the activities of ABL1 inhibited inflammatory signaling. Conclusions ROS-activated ABL1 mediates inflammation through regulating NF-κB1 and STAT3, which subsequently leads to the development of GC and GC-related depression.
Collapse
|
30
|
Li F, Wu Y, Chen L, Hu L, Zhu F, He Q. High iodine induces DNA damage in autoimmune thyroiditis partially by inhibiting the DNA repair protein MTH1. Cell Immunol 2019; 344:103948. [PMID: 31311621 DOI: 10.1016/j.cellimm.2019.103948] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/11/2019] [Accepted: 07/01/2019] [Indexed: 01/06/2023]
Abstract
This study aims to investigate the level of DNA damage in high iodine (HI)-induced autoimmune thyroiditis (AIT), and to explore the role of DNA repair protein MutT homolog-1 (MTH1) in this process. The levels of pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-8 were measured using qRT-PCR and ELISA. The apoptosis was evaluated using TUNEL staining. The pathological changes of thyroid tissues were evaluated using hematoxylin and eosin (HE) staining. The DNA damage was assessed by determining the expression of 8-hydroxy-2'deoxyguanosine (8-OHdG; an indicator of oxidative DNA damage) and performing the Comet assay. Our results showed that both the HI-treated NOD.H-2h4 mice (experimental AIT mice) and the HI-treated mouse thyroid follicular epithelial cells showed enhanced inflammation, apoptosis, and DNA damage level, accompanied by decreased MTH1 expression. Importantly, overexpression of MTH1 effectively abrogated the HI-induced enhancement of inflammation, apoptosis, and DNA damage in mouse thyroid follicular epithelial cells. In conclusion, HI treatment induces DNA damage in AIT, at least in part, by inhibiting the DNA repair protein MTH1.
Collapse
Affiliation(s)
- Fuqiang Li
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Yijun Wu
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Liang Chen
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Liang Hu
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Feng Zhu
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qiwen He
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
31
|
Zhou W, Ma L, Yang J, Qiao H, Li L, Guo Q, Ma J, Zhao L, Wang J, Jiang G, Wan X, Adam Goscinski M, Ding L, Zheng Y, Li W, Liu H, Suo Z, Zhao W. Potent and specific MTH1 inhibitors targeting gastric cancer. Cell Death Dis 2019; 10:434. [PMID: 31164636 PMCID: PMC6547740 DOI: 10.1038/s41419-019-1665-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 01/22/2023]
Abstract
Human mutT homolog 1(MTH1), the oxidized dNTP pool sanitizer enzyme, has been reported to be highly expressed in various malignant tumors. However, the oncogenic role of MTH1 in gastric cancer remains to be determined. In the current study, we found that MTH1 was overexpressed in human gastric cancer tissues and cells. Using an in vitro MTH1 inhibitor screening system, the compounds available in our laboratory were screened and the small molecules containing 5-cyano-6-phenylpyrimidine structure were firstly found to show potently and specifically inhibitory effect on MTH1, especially compound MI-743 with IC50 = 91.44 ± 1.45 nM. Both molecular docking and target engagement experiments proved that MI-743 can directly bind to MTH1. Moreover, MI-743 could not only inhibit cell proliferation in up to 16 cancer cell lines, especially gastric cancer cells HGC-27 and MGC-803, but also significantly induce MTH1-related 8-oxo-dG accumulation and DNA damage. Furthermore, the growth of xenograft tumours derived by injection of MGC-803 cells in nude mice was also significantly inhibited by MI-743 treatment. Importantly, MTH1 knockdown by siRNA in those two gastric cancer cells exhibited the similar findings. Our findings indicate that MTH1 is highly expressed in human gastric cancer tissues and cell lines. Small molecule MI-743 with 5-cyano-6-phenylpyrimidine structure may serve as a novel lead compound targeting the overexpressed MTH1 for gastric cancer treatment.
Collapse
Affiliation(s)
- Wenjuan Zhou
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- Department of Pathology, Oslo University Hospital, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway
| | - Liying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Jing Yang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Hui Qiao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Lingyu Li
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Qian Guo
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Jinlian Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Lijuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Junwei Wang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Guozhong Jiang
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xiangbin Wan
- Department of General Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450001, China
| | - Mariusz Adam Goscinski
- Department of Urology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Lina Ding
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yichao Zheng
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China.
| | - Zhenhe Suo
- Department of Pathology, Oslo University Hospital, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway.
| | - Wen Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
32
|
Zhao H, Pan H, Wang H, Chai P, Ge S, Jia R, Fan X. SKP2 targeted inhibition suppresses human uveal melanoma progression by blocking ubiquitylation of p27. Onco Targets Ther 2019; 12:4297-4308. [PMID: 31213847 PMCID: PMC6549483 DOI: 10.2147/ott.s203888] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/27/2019] [Indexed: 12/25/2022] Open
Abstract
Background: SKP2 is considered an oncogene involved in various malignancies. SKP2 protein is a critical subunit of the SKP1-CUL1-F-box (SCF) E3 ligase complex which affects the cell cycle profoundly by specifically recognizing cell cycle regulators and mediating their ubiquitylation and proteasomal degradation. SKP2 dysfunction is characteristic of many tumor cells. However, its role in uveal melanoma (UM) has not been elucidated. Materials and methods: We analyzed the expressions of SKP2 in different UM cell lines compared with normal pigment cell by RNA-seq, RT-qPCR and Western blot. We then knocked down SKP2 in OM431 and MUM2B cells and confirmed its roles in cell proliferation via CCK8 assay. The sensitivity of cells to SKP2 inhibitor C1 (SKPin C1) in vitro was evaluated by CCK8 assay and colony formation assay, and the sensitivity of MUM2B cells to SKPin C1 in vivo was estimated using the nude mouse-based xenograft model. Western blot and Immunoprecipitation assay were performed to detect the change of p27 and its ubiquitylation level in UM cells treated with SKPin C1, respectively. Results: The results showed that SKP2 was significantly highly expressed in UM cells. SKP2 promoted the progression of UM and knockdown of SKP2 inhibited cell proliferation in UM cells. SKP2 inhibitor C1 that targets SKP2 essentially inhibits the growth of UM cells both in vivo and in vitro. SKP2 inhibitor C1 decreased the degradation of p27 by blocking ubiquitylation of p27, resulting in p27 accumulation and cell cycle arrest in UM cells. Conclusion: Our findings demonstrated that SKP2 targeted inhibition suppresses UM cell proliferation and provides new options and possibilities for targeted therapies in UM.
Collapse
Affiliation(s)
- Hongbo Zhao
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology , Shanghai, People's Republic of China
| | - Hui Pan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology , Shanghai, People's Republic of China
| | - Huixue Wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology , Shanghai, People's Republic of China
| | - Peiwei Chai
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology , Shanghai, People's Republic of China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology , Shanghai, People's Republic of China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology , Shanghai, People's Republic of China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology , Shanghai, People's Republic of China
| |
Collapse
|
33
|
Rai P, Sobol RW. Mechanisms of MTH1 inhibition-induced DNA strand breaks: The slippery slope from the oxidized nucleotide pool to genotoxic damage. DNA Repair (Amst) 2019; 77:18-26. [PMID: 30852368 DOI: 10.1016/j.dnarep.2019.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Unlike normal tissues, tumor cells possess a propensity for genomic instability, resulting from elevated oxidant levels produced by oncogenic signaling and aberrant cellular metabolism. Thus, targeting mechanisms that protect cancer cells from the tumor-inhibitory consequences of their redox imbalance and spontaneous DNA-damaging events is expected to have broad-spectrum efficacy and a high therapeutic index. One critical mechanism for tumor cell protection from oxidant stress is the hydrolysis of oxidized nucleotides. Human MutT homolog 1 (MTH1), the mammalian nudix (nucleoside diphosphate X) pyrophosphatase (NUDT1), protects tumor cells from oxidative stress-induced genomic DNA damage by cleansing the nucleotide pool of oxidized purine nucleotides. Depletion or pharmacologic inhibition of MTH1 results in genomic DNA strand breaks in many cancer cells. However, the mechanisms underlying how oxidized nucleotides, thought mainly to be mutagenic rather than genotoxic, induce DNA strand breaks are largely unknown. Given the recent therapeutic interest in targeting MTH1, a better understanding of such mechanisms is crucial to its successful translation into the clinic and in identifying the molecular contexts under which its inhibition is likely to be beneficial. Here we provide a comprehensive perspective on MTH1 function and its importance in protecting genome integrity, in the context of tumor-associated oxidative stress and the mechanisms that likely lead to irreparable DNA strand breaks as a result of MTH1 inhibition.
Collapse
Affiliation(s)
- Priyamvada Rai
- Department of Medicine/Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States; Sylvester Comprehensive Cancer Center, Miami, FL, 33136, United States.
| | - Robert W Sobol
- Mitchell Cancer Institute, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, United States.
| |
Collapse
|
34
|
Cannavò SP, Tonacci A, Bertino L, Casciaro M, Borgia F, Gangemi S. The role of oxidative stress in the biology of melanoma: A systematic review. Pathol Res Pract 2019; 215:21-28. [DOI: 10.1016/j.prp.2018.11.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022]
|
35
|
Dual functions for OVAAL in initiation of RAF/MEK/ERK prosurvival signals and evasion of p27-mediated cellular senescence. Proc Natl Acad Sci U S A 2018; 115:E11661-E11670. [PMID: 30478051 PMCID: PMC6294934 DOI: 10.1073/pnas.1805950115] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Here, we report that the long noncoding RNA (lncRNA) ovarian adenocarcinoma-amplified lncRNA (OVAAL) is a mediator of cancer cell resistance, counteracting the effects of apoptosis-inducing agents acting through both the extrinsic and intrinsic pathways. Building upon previous reports associating OVAAL amplification with ovarian and endometrial cancers, we now show that OVAAL overexpression occurs during the pathogenesis of colorectal cancer and melanoma. Mechanistically, our findings also establish that OVAAL expression more generally contributes a prosurvival role to cancer cells under steady-state conditions. OVAAL accomplishes these actions utilizing distinct functional modalities: one promoting activation of RAF/MEK/ERK signaling and the other blocking cell entry into senescence. Our study demonstrates that expression of a single OVAAL in cancer cells drives two distinct but coordinated actions contributing to cancer pathology. Long noncoding RNAs (lncRNAs) function through a diverse array of mechanisms that are not presently fully understood. Here, we sought to find lncRNAs differentially regulated in cancer cells resistant to either TNF-related apoptosis-inducing ligand (TRAIL) or the Mcl-1 inhibitor UMI-77, agents that act through the extrinsic and intrinsic apoptotic pathways, respectively. This work identified a commonly up-regulated lncRNA, ovarian adenocarcinoma-amplified lncRNA (OVAAL), that conferred apoptotic resistance in multiple cancer types. Analysis of clinical samples revealed OVAAL expression was significantly increased in colorectal cancers and melanoma in comparison to the corresponding normal tissues. Functional investigations showed that OVAAL depletion significantly inhibited cancer cell proliferation and retarded tumor xenograft growth. Mechanically, OVAAL physically interacted with serine/threonine-protein kinase 3 (STK3), which, in turn, enhanced the binding between STK3 and Raf-1. The ternary complex OVAAL/STK3/Raf-1 enhanced the activation of the RAF protooncogene serine/threonine-protein kinase (RAF)/mitogen-activated protein kinase kinase 1 (MEK)/ERK signaling cascade, thus promoting c-Myc–mediated cell proliferation and Mcl-1–mediated cell survival. On the other hand, depletion of OVAAL triggered cellular senescence through polypyrimidine tract-binding protein 1 (PTBP1)–mediated p27 expression, which was regulated by competitive binding between OVAAL and p27 mRNA to PTBP1. Additionally, c-Myc was demonstrated to drive OVAAL transcription, indicating a positive feedback loop between c-Myc and OVAAL in controlling tumor growth. Taken together, these results reveal that OVAAL contributes to the survival of cancer cells through dual mechanisms controlling RAF/MEK/ERK signaling and p27-mediated cell senescence.
Collapse
|
36
|
La T, Liu GZ, Farrelly M, Cole N, Feng YC, Zhang YY, Sherwin SK, Yari H, Tabatabaee H, Yan XG, Guo ST, Liu T, Thorne RF, Jin L, Zhang XD. A p53-Responsive miRNA Network Promotes Cancer Cell Quiescence. Cancer Res 2018; 78:6666-6679. [PMID: 30301840 DOI: 10.1158/0008-5472.can-18-1886] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/06/2018] [Accepted: 10/02/2018] [Indexed: 11/16/2022]
Abstract
: Cancer cells in quiescence (G0 phase) are resistant to death, and re-entry of quiescent cancer cells into the cell-cycle plays an important role in cancer recurrence. Here we show that two p53-responsive miRNAs utilize distinct but complementary mechanisms to promote cancer cell quiescence by facilitating stabilization of p27. Purified quiescent B16 mouse melanoma cells expressed higher levels of miRNA-27b-3p and miRNA-455-3p relative to their proliferating counterparts. Induction of quiescence resulted in increased levels of these miRNAs in diverse types of human cancer cell lines. Inhibition of miRNA-27b-3p or miRNA-455-3p reduced, whereas its overexpression increased, the proportion of quiescent cells in the population, indicating that these miRNAs promote cancer cell quiescence. Accordingly, cancer xenografts bearing miRNA-27b-3p or miRNA-455-3p mimics were retarded in growth. miRNA-27b-3p targeted cyclin-dependent kinase regulatory subunit 1 (CKS1B), leading to reduction in p27 polyubiquitination mediated by S-phase kinase-associated protein 2 (Skp2). miRNA-455-3p targeted CDK2-associated cullin domain 1 (CAC1), which enhanced CDK2-mediated phosphorylation of p27 necessary for its polyubiquitination. Of note, the gene encoding miRNA-27b-3p was embedded in the intron of the chromosome 9 open reading frame 3 gene that was transcriptionally activated by p53. Similarly, the host gene of miRNA-455-3p, collagen alpha-1 (XXVII) chain, was also a p53 transcriptional target. Collectively, our results identify miRNA-27b-3p and miRNA-455-3p as important regulators of cancer cell quiescence in response to p53 and suggest that manipulating miRNA-27b-3p and miRNA-455-3p may constitute novel therapeutic avenues for improving outcomes of cancer treatment. SIGNIFICANCE: Two novel p53-responsive microRNAs whose distinct mechanisms of action both stabilize p27 to promote cell quiescence and may serve as therapeutic avenues for improving outcomes of cancer treatment.
Collapse
Affiliation(s)
- Ting La
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Guang Zhi Liu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Henan, China
| | - Margaret Farrelly
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Nicole Cole
- Research Infrastructure, Research and Innovation Division, The University of Newcastle, New South Wales, Australia
| | - Yu Chen Feng
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Yuan Yuan Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Simonne K Sherwin
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Hamed Yari
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Hessam Tabatabaee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Xu Guang Yan
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Su Tang Guo
- Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Shanxi, China
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, New South Wales, Australia
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Henan, China.,School of Environmental and Life Sciences, University of Newcastle, New South Wales, Australia
| | - Lei Jin
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia.
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia. .,Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Henan, China
| |
Collapse
|
37
|
Zhang D, Lin J, Chao Y, Zhang L, Jin L, Li N, He R, Ma B, Zhao W, Han C. Regulation of the adaptation to ER stress by KLF4 facilitates melanoma cell metastasis via upregulating NUCB2 expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:176. [PMID: 30055641 PMCID: PMC6064624 DOI: 10.1186/s13046-018-0842-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/13/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Adaptation to ER stress has been indicated to play an important role in resistance to therapy in human melanoma. However, the relationship between adaptation to ER stress and cell metastasis in human melanoma remains unclear. METHODS The relationship of adaptation to ER stress and cell metastasis was investigated using transwell and mouse metastasis assays. The potential molecular mechanism of KLF4 in regulating the adaptation to ER stress and cell metastasis was investigated using RNA sequencing analysis, q-RT-PCR and western blot assays. The transcriptional regulation of nucleobindin 2 (NUCB2) by KLF4 was identified using bioinformatic analysis, luciferase assay, and chromatin immunoprecipitation (ChIP). The clinical significance of KLF4 and NUCB2 was based on human tissue microarray (TMA) analysis. RESULTS Here, we demonstrated that KLF4 was induced by ER stress in melanoma cells, and increased KLF4 inhibited cell apoptosis and promoted cell metastasis. Further mechanistic studies revealed that KLF4 directly bound to the promoter of NUCB2, facilitating its transcription. Additionally, an increase in KLF4 promoted melanoma ER stress resistance, tumour growth and cell metastasis by regulating NCUB2 expression in vitro and in vivo. Elevated KLF4 was found in human melanoma tissues, which was associated with NUCB2 expression. CONCLUSION Our data revealed that the promotion of ER stress resistance via the KLF4-NUCB2 axis is essential for melanoma cell metastasis, and KLF4 may be a promising specific target for melanoma therapy.
Collapse
Affiliation(s)
- Dongmei Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.,Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Jingrong Lin
- Department of Dermatology, the First Affiliated Hospital, Dalian Medical University, Liaoning, 116027, China
| | - Yulin Chao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Lu Zhang
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China
| | - Lei Jin
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Na Li
- Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ruiping He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Binbin Ma
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China
| | - Wenzhi Zhao
- Department of Orthopedics, Second Affiliated Hospital, Dalian Medical University, Dalian, 116044, China.
| | - Chuanchun Han
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
38
|
Mansour MA. Ubiquitination: Friend and foe in cancer. Int J Biochem Cell Biol 2018; 101:80-93. [PMID: 29864543 DOI: 10.1016/j.biocel.2018.06.001] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/05/2023]
Abstract
Dynamic modulation and posttranslational modification of proteins are tightly controlled biological processes that occur in response to physiological cues. One such dynamic modulation is ubiquitination, which marks proteins for degradation via the proteasome, altering their localization, affecting their activity, and promoting or interfering with protein interactions. Hence, ubiquitination is crucial for a plethora of physiological processes, including cell survival, differentiation and innate and adaptive immunity. Similar to kinases, components of the ubiquitination system are often deregulated, leading to a variety of diseases, such as cancer and neurodegenerative disorders. In a context-dependent manner, ubiquitination can regulate both tumor-suppressing and tumor-promoting pathways in cancer. This review outlines how components of the ubiquitination systems (e.g. E3 ligases and deubiquitinases) act as oncogenes or tumor suppressors according to the nature of their substrates. Furthermore, I interrogate how the current knowledge of the differential roles of ubiquitination in cancer lead to technical advances to inhibit or reactivate the components of the ubiquitination system accordingly.
Collapse
Affiliation(s)
- Mohammed A Mansour
- Institute of Cancer Sciences, University of Glasgow, United Kingdom; The CRUK Beatson Institute, Glasgow, Switchback Road, G61 1BD, United Kingdom; Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
39
|
Lin J, Zhang D, Fan Y, Chao Y, Chang J, Li N, Han L, Han C. Regulation of Cancer Stem Cell Self-Renewal by HOXB9 Antagonizes Endoplasmic Reticulum Stress-Induced Melanoma Cell Apoptosis via the miR-765-FOXA2 Axis. J Invest Dermatol 2018; 138:1609-1619. [PMID: 29408459 DOI: 10.1016/j.jid.2018.01.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/13/2018] [Accepted: 01/15/2018] [Indexed: 01/13/2023]
Abstract
Adaptation to endoplasmic reticulum (ER) stress has been indicated as a driver of malignancy and resistance to therapy in human melanoma. However, the relationship between cancer stem cells and adaptation to ER stress remains unclear. Here, we show that the ratio of cancer stem cells is increased in ER stress-resistant melanoma cells, which inhibit ER stress-induced apoptosis and promote tumorigenesis. Further mechanistic studies showed that HOXB9 triggered by ER stress favors cancer stem cell self-renewal and enhances ER stress resistance. HOXB9 directly binds to the promoter of microRNA-765 and facilitates its transcription, which in turn targets FOXA2, resulting in a FOXA2 decrease and cancer stem cell increase. Additionally, an increase in HOXB9 promotes melanoma growth and inhibits cell apoptosis in a mouse xenograft model. Elevated HOXB9 is found in human melanoma tissues, which is associated with microRNA-765 up-regulation and FOXA2 decreases. Thus, our data showed that the HOXB9-dependent, microRNA-765-mediated FOXA2 pathway contributes to the survival of melanoma under ER stress by maintaining the properties of cancer stem cells.
Collapse
Affiliation(s)
- Jingrong Lin
- Institute of Cancer Stem Cell and Department of Dermatology of the First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Dongmei Zhang
- Department of Physiology of College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Yongsheng Fan
- Institute of Cancer Stem Cell and Department of Dermatology of the First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Yulin Chao
- Institute of Cancer Stem Cell and Department of Dermatology of the First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Jinming Chang
- Institute of Cancer Stem Cell and Department of Dermatology of the First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Na Li
- Department of Physiology of College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Linlin Han
- Physical Examination Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chuanchun Han
- Institute of Cancer Stem Cell and Department of Dermatology of the First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|