1
|
Sun H, Meng Y, Yao L, Du S, Li Y, Zhou Q, Liu Y, Dian Y, Sun Y, Wang X, Liang X, Deng G, Chen X, Zeng F. Ubiquitin-specific protease 22 controls melanoma metastasis and vulnerability to ferroptosis through targeting SIRT1/PTEN/PI3K signaling. MedComm (Beijing) 2024; 5:e684. [PMID: 39135915 PMCID: PMC11318338 DOI: 10.1002/mco2.684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 08/15/2024] Open
Abstract
Metastasis is a major contributing factor that affects the prognosis of melanoma patients. Nevertheless, the underlying molecular mechanisms involved in melanoma metastasis are not yet entirely understood. Here, we identified ubiquitin-specific protease 22 (USP22) as a pro-oncogenic protein in melanoma through screening the survival profiles of 52 ubiquitin-specific proteases (USPs). USP22 demonstrates a strong association with poor clinical outcomes and is significantly overexpressed in melanoma. Ablation of USP22 expression remarkably attenuates melanoma migration, invasion, and epithelial-mesenchymal transition in vitro and suppresses melanoma metastasis in vivo. Mechanistically, USP22 controls melanoma metastasis through the SIRT1/PTEN/PI3K pathway. In addition, we conducted an United States Food and Drug Administration-approved drug library screening and identified topotecan as a clinically applicable USP22-targeting molecule by promoting proteasomal degradation of USP22. Finally, we found that both pharmacological and genetic silence of USP22 sensitize RSL3-induced ferroptosis through suppressing the PI3K/Akt/mTOR pathway and its downstream SCD, and ferroptosis inhibitor could partly rescued the decreased lung metastasis by topotecan in vivo. Overall, our findings reveal a prometastatic role of USP22 and identify topotecan as a potent USP22-targeting drug to limit melanoma metastasis.
Collapse
Affiliation(s)
- Huiyan Sun
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
- Department of Breast ReconstructionTianjin Medical UniversityCancer Institute and HospitalTianjinChina
| | - Yu Meng
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Lei Yao
- Department of Liver SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Songtao Du
- Department of Colorectal Surgical OncologyThe Tumor Hospital of Harbin Medical UniversityHarbinChina
| | - Yayun Li
- Department of DermatologyThe Third Xiangya Hospital Central South UniversityChangshaChina
| | - Qian Zhou
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yihuang Liu
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yating Dian
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yuming Sun
- Department of Plastic and Cosmetic SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Xiaomin Wang
- Department of Breast SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Xiao‐wei Liang
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Guangtong Deng
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Xiang Chen
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Furong Zeng
- Department of OncologyXiangya Hospital Central South UniversityChangshaChina
| |
Collapse
|
2
|
Liu J, Fu J, Fu P, Liu M, Liu Z, Song B. Pitavastatin sensitizes the EGFR-TKI associated resistance in lung cancer by inhibiting YAP/AKT/BAD-BCL-2 pathway. Cancer Cell Int 2024; 24:224. [PMID: 38943199 PMCID: PMC11214206 DOI: 10.1186/s12935-024-03416-z] [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: 10/17/2023] [Accepted: 06/22/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Despite effective strategies, resistance in EGFR mutated lung cancer remains a challenge. Metabolic reprogramming is one of the main mechanisms of tumor drug resistance. A class of drugs known as "statins" inhibit lipid cholesterol metabolism and are widely used in patients with cardiovascular diseases. Previous studies have also documented its ability to improve the therapeutic impact in lung cancer patients who receive EGFR-TKI therapy. Therefore, the effect of statins on targeted drug resistance to lung cancer remains to be investigated. METHODS Prolonged exposure to gefitinib resulted in the emergence of a resistant lung cancer cell line (PC9GR) from the parental sensitive cell line (PC9), which exhibited a traditional EGFR mutation. The CCK-8 assay was employed to assess the impact of various concentrations of pitavastatin on cellular proliferation. RNA sequencing was conducted to detect differentially expressed genes and their correlated pathways. For the detection of protein expression, Western blot was performed. The antitumor activity of pitavastatin was evaluated in vivo via a xenograft mouse model. RESULTS PC9 gefitinib resistant strains were induced by low-dose maintenance. Cell culture and animal-related studies validated that the application of pitavastatin inhibited the proliferation of lung cancer cells, promoted cell apoptosis, and restrained the acquired resistance to EGFR-TKIs. KEGG pathway analysis showed that the hippo/YAP signaling pathway was activated in PC9GR cells relative to PC9 cells, and the YAP expression was inhibited by pitavastatin administration. With YAP RNA interference, pAKT, pBAD and BCL-2 expression was decreased, while BAX expression as increased. Accordingly, YAP down-regulated significantly increased apoptosis and decreased the survival rate of gefitinib-resistant lung cancer cells. After pAKT was increased by SC79, apoptosis of YAP down-regulated cells induced by gefitinib was decreased, and the cell survival rate was increased. Mechanistically, these effects of pitavastatin are associated with the YAP pathway, thereby inhibiting the downstream AKT/BAD-BCL-2 signaling pathway. CONCLUSION Our study provides a molecular basis for the clinical application of the lipid-lowering drug pitavastatin enhances the susceptibility of lung cancer to EGFR-TKI drugs and alleviates drug resistance.
Collapse
Affiliation(s)
- Jie Liu
- Cancer Center, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Jialei Fu
- Shandong Academy of Chinese Medicine, Jinan, China
| | - Ping Fu
- Department of Chemotherapy, Jinan Zhangqiu District People's Hospital, Jinan, China
| | - Menghan Liu
- Clinical Medical College, Shandong First Medical University, Jinan, China
| | - Zining Liu
- Department of Nuclear Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China.
| | - Bao Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
3
|
Liu J, Wang Y, Zhang S, Sun L, Shi Y. ADAM9 deubiquitination induced by USP22 suppresses proliferation, migration, invasion, and epithelial-mesenchymal transition of trophoblast cells in preeclampsia. Placenta 2024; 146:50-57. [PMID: 38176298 DOI: 10.1016/j.placenta.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024]
Abstract
INTRODUCTION The dysregulation of deubiquitination has been shown to affect the development of pre-eclampsia (PE). A disintegrin and metalloprotease 9 (ADAM9) plays roles in diverse physiological contexts, including PE. Here, this study aimed to investigate whether ADAM9 regulated trophoblast cell dysfunction through ubiquitin-specific protease 22 (USP22) deubiquitinase-mediated deubiquitination during PE. METHODS Levels of genes and proteins were tested via qRT-PCR and western blotting assays. Cell proliferation, migration, and invasion were detected using cell counting kit-8, 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell and wound healing assays, respectively. Epithelial-mesenchymal transition related markers were assayed using western blotting. Proteins between USP22 and ADAM9 were identified by co-immunoprecipitation assay. RESULTS ADAM9 was highly expressed in PE patients, functionally, ADAM9 overexpression weakened the proliferation, migration, invasion, and EMT progression in trophoblast cells. Mechanistically, the deubiquitinase USP22 removed ubiquitination on ADAM9 and maintained its stability. Forced expression of USP22 also suppressed the proliferation and mobility in trophoblast cells. Moreover, the regulatory effects of USP22 on trophoblast cells were reversed by ADAM9 silencing. In addition, USP22 interacted with ADAM9 to regulate the activation of Wnt/β-catenin pathway. DISCUSSION ADAM9 was deubiquitinated and stabilized by USP22 and then suppressed the proliferation, migration, invasion, and EMT progression in trophoblast cells, indicating a new pathway of USP10/RUNX1 axis in PE process.
Collapse
Affiliation(s)
- Jie Liu
- Department of Gynecology and Obstetrics, Yantaishan Hospital, Yantai City, 264001, Shandong, China
| | - Yan Wang
- Department of Gynecology and Obstetrics, Yantaishan Hospital, Yantai City, 264001, Shandong, China
| | - Suqin Zhang
- Department of Gynecology and Obstetrics, Yantaishan Hospital, Yantai City, 264001, Shandong, China
| | - Liyan Sun
- Department of Pharmacy, Yantaishan Hospital, Yantai City, 264001, Shandong, China
| | - Yanmei Shi
- Department of Gynecology and Obstetrics, Yantaishan Hospital, Yantai City, 264001, Shandong, China.
| |
Collapse
|
4
|
Ma ZR, Xiong QW, Cai SZ, Ding LT, Yin CH, Xia HL, Liu W, Dai S, Zhang Y, Zhu ZH, Huang ZJ, Wang Q, Yan XM. USP18 enhances the resistance of BRAF-mutated melanoma cells to vemurafenib by stabilizing cGAS expression to induce cell autophagy. Int Immunopharmacol 2023; 122:110617. [PMID: 37478666 DOI: 10.1016/j.intimp.2023.110617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/14/2023] [Accepted: 07/04/2023] [Indexed: 07/23/2023]
Abstract
This study aims to discern the possible molecular mechanism of the effect of ubiquitin-specific peptidase 18 (USP18) on the resistance to BRAF inhibitor vemurafenib in BRAF V600E mutant melanoma by regulating cyclic GMP-AMP synthase (cGAS). The cancer tissues of BRAF V600E mutant melanoma patients before and after vemurafenib treatment were collected, in which the protein expression of USP18 and cGAS was determined. A BRAF V600E mutant human melanoma cell line (A2058R) resistant to vemurafenib was constructed with its viability, apoptosis, and autophagy detected following overexpression and depletion assays of USP18 and cGAS. Xenografted tumors were transplanted into nude mice for in vivo validation. Bioinformatics analysis showed that the expression of cGAS was positively correlated with USP18 in melanoma, and USP18 was highly expressed in melanoma. The expression of cGAS and USP18 was up-regulated in cancer tissues of vemurafenib-resistant patients with BRAF V600E mutant melanoma. Knockdown of cGAS inhibited the resistance to vemurafenib in A2058R cells and the protective autophagy induced by vemurafenib in vitro. USP18 could deubiquitinate cGAS to promote its protein stability. In vivo experimentations confirmed that USP18 promoted vemurafenib-induced protective autophagy by stabilizing cGAS protein, which promoted resistance to vemurafenib in BRAF V600E mutant melanoma cells. Collectively, USP18 stabilizes cGAS protein expression through deubiquitination and induces autophagy of melanoma cells, thereby promoting the resistance to vemurafenib in BRAF V600E mutant melanoma.
Collapse
Affiliation(s)
- Zhou-Rui Ma
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou 215025, PR China; Suzhou Key Laboratory of Congenital Structural Deformities, Suzhou 215025, Jiangsu, PR China
| | - Qian-Wei Xiong
- Suzhou Key Laboratory of Congenital Structural Deformities, Suzhou 215025, Jiangsu, PR China; Department of Urology, Children's Hospital of Soochow University, Suzhou 215025, PR China
| | - Shi-Zhong Cai
- Suzhou Key Laboratory of Congenital Structural Deformities, Suzhou 215025, Jiangsu, PR China; Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou 215025, PR China
| | - Ling-Tao Ding
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214000, PR China
| | - Chao-Hong Yin
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214000, PR China
| | - Hong-Liang Xia
- Suzhou Key Laboratory of Congenital Structural Deformities, Suzhou 215025, Jiangsu, PR China; Department of Urology, Children's Hospital of Soochow University, Suzhou 215025, PR China
| | - Wei Liu
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou 215025, PR China
| | - Shu Dai
- Suzhou Key Laboratory of Congenital Structural Deformities, Suzhou 215025, Jiangsu, PR China
| | - Yue Zhang
- Soochow University, Suzhou 215006, PR China
| | - Zhen-Hong Zhu
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou 215025, PR China
| | - Zhi-Jian Huang
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou 215025, PR China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou 215025, PR China.
| | - Xiang-Ming Yan
- Suzhou Key Laboratory of Congenital Structural Deformities, Suzhou 215025, Jiangsu, PR China; Department of Surgery, Children's Hospital of Soochow University, Suzhou 215025, PR China.
| |
Collapse
|
5
|
Wu L, Ou Z, Liu P, Zhao C, Tong S, Wang R, Li Y, Yuan J, Chen M, Fan B, Zu X, Wang Y, Tang J. ATXN3 promotes prostate cancer progression by stabilizing YAP. Cell Commun Signal 2023; 21:152. [PMID: 37349820 PMCID: PMC10286397 DOI: 10.1186/s12964-023-01073-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/11/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Prostate cancer (PC) is the most common neoplasm and is the second leading cause of cancer-related deaths in men worldwide. The Hippo tumor suppressor pathway is highly conserved in mammals and plays an important role in carcinogenesis. YAP is one of major key effectors of the Hippo pathway. However, the mechanism supporting abnormal YAP expression in PC remains to be characterized. METHODS Western blot was used to measure the protein expression of ATXN3 and YAP, while the YAP target genes were measured by real-time PCR. CCK8 assay was used to detect cell viability; transwell invasion assay was used to measure the invasion ability of PC. The xeno-graft tumor model was used for in vivo study. Protein stability assay was used to detect YAP protein degradation. Immuno-precipitation assay was used to detect the interaction domain between YAP and ATXN3. The ubiquitin-based Immuno-precipitation assays were used to detect the specific ubiquitination manner happened on YAP. RESULTS In the present study, we identified ATXN3, a DUB enzyme in the ubiquitin-specific proteases family, as a bona fide deubiquitylase of YAP in PC. ATXN3 was shown to interact with, deubiquitylate, and stabilize YAP in a deubiquitylation activity-dependent manner. Depletion of ATXN3 decreased the YAP protein level and the expression of YAP/TEAD target genes in PC, including CTGF, ANKRD1 and CYR61. Further mechanistic study revealed that the Josephin domain of ATXN3 interacted with the WW domain of YAP. ATXN3 stabilized YAP protein via inhibiting K48-specific poly-ubiquitination process on YAP protein. In addition, ATXN3 depletion significantly decreased PC cell proliferation, invasion and stem-like properties. The effects induced by ATXN3 depletion could be rescued by further YAP overexpression. CONCLUSIONS In general, our findings establish a previously undocumented catalytic role for ATXN3 as a deubiquitinating enzyme of YAP and provides a possible target for the therapy of PC. Video Abstract.
Collapse
Affiliation(s)
- Longxiang Wu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenyu Ou
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Peihua Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Zhao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Shiyu Tong
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Ruizhe Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Yangle Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Junbin Yuan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Minfeng Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Benyi Fan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongjie Wang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China.
| | - Jianing Tang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
6
|
Jia X, He L, Yang Z. Recent advances in the role of Yes-associated protein in dermatosis. Skin Res Technol 2023; 29:e13285. [PMID: 36973973 PMCID: PMC10155855 DOI: 10.1111/srt.13285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 03/04/2023]
Abstract
BACKGROUND Dermatosis is a general term for diseases of the skin and skin appendages including scleroderma, psoriasis, bullous disease, atopic dermatitis, basal cell carcinoma, squamous cell carcinoma, and melanoma. These diseases affect millions of individuals globally and are a serious public health concern. However, the pathogenesis of skin diseases is not fully understood, and treatments are not optimal. Yes-associated protein (YAP) is a transcriptional coactivator that plays a role in the regulation of gene transcription and signal transduction. AIMS To study the role of Yes-associated protein in skin diseases. MATERIALS AND METHODS The present review summarizes recent advances in our understanding of the role of YAP in skin diseases, current treatments that target YAP, and potential avenues for novel therapies. RESULTS Abnormal YAP expression has been implicated in occurrence and development of dermatosis. YAP regulates the cell homeostasis, proliferation, differentiation, apoptosis, angiopoiesis, and epithelial-to-mesenchymal transition, among other processes. As well as, it serves as a potential target in many biological processes for treating dermatosis. CONCLUSIONS The effects of YAP on the skin are complex and require multidimensional investigational approaches. YAP functions as an oncoprotein that can promote the occurrence and development of cancer, but there is currently limited information on the therapeutic potential of YAP inhibition for cancer treatment. Additional studies are also needed to clarify the role of YAP in the development and maturation of dermal fibroblasts; skin barrier function, homeostasis, aging, and melanin production; and dermatosis.
Collapse
Affiliation(s)
- Xiaorong Jia
- Department of DermatologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Li He
- Department of DermatologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Zhi Yang
- Department of DermatologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| |
Collapse
|
7
|
Ming H, Li B, Jiang J, Qin S, Nice EC, He W, Lang T, Huang C. Protein degradation: expanding the toolbox to restrain cancer drug resistance. J Hematol Oncol 2023; 16:6. [PMID: 36694209 PMCID: PMC9872387 DOI: 10.1186/s13045-023-01398-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/01/2023] [Indexed: 01/25/2023] Open
Abstract
Despite significant progress in clinical management, drug resistance remains a major obstacle. Recent research based on protein degradation to restrain drug resistance has attracted wide attention, and several therapeutic strategies such as inhibition of proteasome with bortezomib and proteolysis-targeting chimeric have been developed. Compared with intervention at the transcriptional level, targeting the degradation process seems to be a more rapid and direct strategy. Proteasomal proteolysis and lysosomal proteolysis are the most critical quality control systems responsible for the degradation of proteins or organelles. Although proteasomal and lysosomal inhibitors (e.g., bortezomib and chloroquine) have achieved certain improvements in some clinical application scenarios, their routine application in practice is still a long way off, which is due to the lack of precise targeting capabilities and inevitable side effects. In-depth studies on the regulatory mechanism of critical protein degradation regulators, including E3 ubiquitin ligases, deubiquitylating enzymes (DUBs), and chaperones, are expected to provide precise clues for developing targeting strategies and reducing side effects. Here, we discuss the underlying mechanisms of protein degradation in regulating drug efflux, drug metabolism, DNA repair, drug target alteration, downstream bypass signaling, sustaining of stemness, and tumor microenvironment remodeling to delineate the functional roles of protein degradation in drug resistance. We also highlight specific E3 ligases, DUBs, and chaperones, discussing possible strategies modulating protein degradation to target cancer drug resistance. A systematic summary of the molecular basis by which protein degradation regulates tumor drug resistance will help facilitate the development of appropriate clinical strategies.
Collapse
Affiliation(s)
- Hui Ming
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Bowen Li
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Jingwen Jiang
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Siyuan Qin
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Weifeng He
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Army Military Medical University, Chongqing, 400038, China.
| | - Tingyuan Lang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, 400030, People's Republic of China. .,Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People's Republic of China.
| | - Canhua Huang
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
8
|
Elu N, Presa N, Mayor U. RNAi-Based Screening for the Identification of Specific Substrate-Deubiquitinase Pairs. Methods Mol Biol 2023; 2602:95-105. [PMID: 36446969 DOI: 10.1007/978-1-0716-2859-1_7] [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] [Indexed: 06/16/2023]
Abstract
Ubiquitination signals are regulated in time and space due to the coordinated action of E3s and DUBs, which enables the precise control of cellular function and homeostasis. Mutations in all types of ubiquitin-proteasome system (UPS) components are related to pathological conditions. The identification of E3/DUBs' ubiquitinated substrates can provide a clearer view of the molecular mechanisms underlying those diseases. However, the analysis of ubiquitinated proteins is not trivial. Here, we propose a protocol to identify DUB/substrate pairs, by combining DUB silencing, specific pull-down of the substrate, and image analysis of its ubiquitinated fraction.
Collapse
Affiliation(s)
- Nagore Elu
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Natalia Presa
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
9
|
Elu N, Osinalde N, Ramirez J, Presa N, Rodriguez JA, Prieto G, Mayor U. Identification of substrates for human deubiquitinating enzymes (DUBs): An up-to-date review and a case study for neurodevelopmental disorders. Semin Cell Dev Biol 2022; 132:120-131. [PMID: 35042675 DOI: 10.1016/j.semcdb.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 12/15/2022]
Abstract
Similar to the reversal of kinase-mediated protein phosphorylation by phosphatases, deubiquitinating enzymes (DUBs) oppose the action of E3 ubiquitin ligases and reverse the ubiquitination of proteins. A total of 99 human DUBs, classified in 7 families, allow in this way for a precise control of cellular function and homeostasis. Ubiquitination regulates a myriad of cellular processes, and is altered in many pathological conditions. Thus, ubiquitination-regulating enzymes are increasingly regarded as potential candidates for therapeutic intervention. In this context, given the predicted easier pharmacological control of DUBs relative to E3 ligases, a significant effort is now being directed to better understand the processes and substrates regulated by each DUB. Classical studies have identified specific DUB substrate candidates by traditional molecular biology techniques in a case-by-case manner. Lately, single experiments can identify thousands of ubiquitinated proteins at a specific cellular context and narrow down which of those are regulated by a given DUB, thanks to the development of new strategies to isolate and enrich ubiquitinated material and to improvements in mass spectrometry detection capabilities. Here we present an overview of both types of studies, discussing the criteria that, in our view, need to be fulfilled for a protein to be considered as a high-confidence substrate of a given DUB. Applying these criteria, we have manually reviewed the relevant literature currently available in a systematic manner, and identified 650 high-confidence substrates of human DUBs. We make this information easily accessible to the research community through an updated version of the DUBase website (https://ehubio.ehu.eus/dubase/). Finally, in order to illustrate how this information can contribute to a better understanding of the physiopathological role of DUBs, we place a special emphasis on a subset of these enzymes that have been associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nagore Elu
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Nerea Osinalde
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Juanma Ramirez
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Natalia Presa
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Jose Antonio Rodriguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Gorka Prieto
- Department of Communications Engineering, University of the Basque Country (UPV/EHU), Bilbao 48013, Spain
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain; Ikerbasque, Basque Foundation for Science, Bilbao 48013, Spain.
| |
Collapse
|
10
|
Targeting the USP7/RRM2 axis drives senescence and sensitizes melanoma cells to HDAC/LSD1 inhibitors. Cell Rep 2022; 40:111396. [PMID: 36130505 DOI: 10.1016/j.celrep.2022.111396] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 07/01/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Deubiquitinating enzymes are key regulators of the ubiquitin-proteasome system and cell cycle, and their dysfunction leads to tumorigenesis. Our in vivo drop-out screens in patient-derived xenograft models identify USP7 as a regulator of melanoma. We show that USP7 downregulation induces cellular senescence, arresting melanoma growth in vivo and proliferation in vitro in BRAF- and NRAS-mutant melanoma. We provide a comprehensive understanding of targets and networks affected by USP7 depletion by performing a global transcriptomic and proteomics analysis. We show that RRM2 is a USP7 target and is regulated by USP7 during S phase of the cell cycle. Ectopic expression of RRM2 in USP7-depleted cells rescues the senescent phenotype. Pharmacological inhibition of USP7 by P5091 phenocopies the shUSP7-induced senescent phenotype. We show that the bifunctional histone deacetylase (HDAC)/LSD1 inhibitor domatinostat has an additive antitumor effect, eliminating P5091-induced senescent cells, paving the way to a therapeutic combination for individuals with melanoma.
Collapse
|
11
|
Ohanna M, Biber P, Deckert M. Emerging Role of Deubiquitinating Enzymes (DUBs) in Melanoma Pathogenesis. Cancers (Basel) 2022; 14:3371. [PMID: 35884430 PMCID: PMC9322030 DOI: 10.3390/cancers14143371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Metastatic melanoma is the leading cause of death from skin cancer. Therapies targeting the BRAF oncogenic pathway and immunotherapies show remarkable clinical efficacy. However, these treatments are limited to subgroups of patients and relapse is common. Overall, the majority of patients require additional treatments, justifying the development of new therapeutic strategies. Non-genetic and genetic alterations are considered to be important drivers of cellular adaptation mechanisms to current therapies and disease relapse. Importantly, modification of the overall proteome in response to non-genetic and genetic events supports major cellular changes that are required for the survival, proliferation, and migration of melanoma cells. However, the mechanisms underlying these adaptive responses remain to be investigated. The major contributor to proteome remodeling involves the ubiquitin pathway, ubiquitinating enzymes, and ubiquitin-specific proteases also known as DeUBiquitinases (DUBs). In this review, we summarize the current knowledge regarding the nature and roles of the DUBs recently identified in melanoma progression and therapeutic resistance and discuss their potential as novel sources of vulnerability for melanoma therapy.
Collapse
Affiliation(s)
- Mickael Ohanna
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| | - Pierric Biber
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| | - Marcel Deckert
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| |
Collapse
|