1
|
Zhuang D, Kang J, Luo H, Tian Y, Liu X, Shao C. ARv7 promotes the escape of prostate cancer cells from androgen deprivation therapy-induced senescence by mediating the SKP2/p27 axis. BMC Biol 2025; 23:66. [PMID: 40022149 PMCID: PMC11871636 DOI: 10.1186/s12915-025-02172-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Androgen deprivation therapy (ADT) induces cellular senescence and tumor stasis, thus serving as the standard treatment for prostate cancer (PCa). However, continuous suppression of canonical androgen receptor signaling actually leads to the switch from androgen-responsive growth to androgen-independent growth, contributing to "escape" from this ADT-induced senescence (AIS) and, subsequently, the development of castration-resistant prostate cancer (CRPC). Unfortunately, the mechanism underlying this phenomenon remains elusive. RESULTS In this study, we demonstrated that androgen receptor splicing variant 7 (ARv7), a dominant factor mediating abnormal AR signaling and ADT resistance, is closely associated with outgrowth from AIS of PCa cells. Mechanistically, ARv7 binds to the promoter of SKP2, activating its transcription, and then promotes the proteasomal degradation of the cell cycle regulator p27 and G1/S transition. In addition, we applied bioinformatic and in vitro analyses to show that SKP2 expression level is dramatically inhibited upon ADT, but its reactivation is one key step during the establishment of CRPC. Finally, we also demonstrated that SKP2 inhibitor treatment can significantly inhibit the growth of androgen-independent cell lines and enhance the efficacy of ADT. CONCLUSIONS Our work reveals a novel role of ARv7 in regulating AIS and suggests that targeting the ARv7/SKP2/p27 axis could be a potential strategy to delay disease progression to the CRPC state during prolonged ADT.
Collapse
Affiliation(s)
- Dian Zhuang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Jinsong Kang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Haoge Luo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yu Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Xiaoping Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Chen Shao
- Department of Biochemistry & Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Zeng J, Chen Z, He Y, Jiang Z, Zhang Y, Dong Q, Chen L, Deng S, He Z, Li L, Li J, Shi J. A patent review of SCF E3 ligases inhibitors for cancer:Structural design, pharmacological activities and structure-activity relationship. Eur J Med Chem 2024; 278:116821. [PMID: 39232359 DOI: 10.1016/j.ejmech.2024.116821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Currently, as the largest family of E3 ubiquitin ligases, Skp1-Cullin 1-F-box (SCF) E3 ligase complexes have attracted extensive attention. Among SCF complexes, Skp2, β-TrCP, and FBXW7 have undergone extensive research on their structures and functions. Previous studies suggest Skp2, β-TrCP, and FBXW7 are overexpressed in numerous cancers. Thus, the SCF E3 ligase complex has become a significant target for the development of anti-cancer drugs. Over the past few decades, a variety of anti-tumor inhibitors targeting the SCF E3 ligase complex have been attempted. However, since almost none of the SCF E3 ligase inhibitors passed clinical trials, the design and synthesis of the new inhibitors are needed. Here, we will introduce the structure and function of Skp2, β-TrCP, and FBXW7, their connections with cancer development, the relevant in vitro and in vivo activities, selectivity, structure-activity relationships, and the therapeutic or preventive application of small molecule inhibitors targeting these three F-box proteins reported in the patent (2010-present). This information will help develop drugs targeting the SCF E3 ubiquitin ligase, providing new strategies for future cancer treatments.
Collapse
Affiliation(s)
- Jing Zeng
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yuxin He
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Zhongliang Jiang
- Hematology Department, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Yi Zhang
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Qin Dong
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Liping Chen
- School of Comprehensive Health Management, Xihua University, Chengdu, 610039, China
| | - Sichun Deng
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Ziyou He
- School of Economics and Management, The University of Hong Kong, Hong Kong, 999077, China
| | - Ling Li
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China; Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, Sichuan, 611137, China.
| | - Jinqi Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
3
|
Khan AQ, Al-Tamimi M, Anver R, Agha MV, Anamangadan G, Raza SS, Ahmad F, Ahmad A, Alam M, Buddenkotte J, Steinhoff M, Uddin S. Targeting of S-phase kinase associated protein 2 stabilized tumor suppressors leading to apoptotic cell death in squamous skin cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167286. [PMID: 38866114 DOI: 10.1016/j.bbadis.2024.167286] [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: 11/21/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
S-phase kinase-associated protein 2 (Skp2) is an F-box protein overexpressed in human cancers and linked with poor prognosis. It triggers cancer pathogenesis, including stemness and drug resistance. In this study, we have explored the potential role of Skp2 targeting in restoring the expression of tumor suppressors in human cutaneous squamous cell carcinoma (cSCC) cells. Our results showed that genetic and pharmacological Skp2 targeting markedly suppressed cSCC cell proliferation, colony growth, spheroid formation, and enhanced sensitization to chemotherapeutic drugs. Further, western blot results demonstrated restoration of tumor suppressor (KLF4) and CDKI (p21) and suppression of vimentin and survivin in Skp2-knocked-down cSCC cells. Importantly, we also explored that Skp2 targeting potentiates apoptosis of cSCC cells through MAPK signaling. Moreover, co-targeting of Skp2 and PI3K/AKT resulted in increased cancer cell death. Interestingly, curcumin, a well-known naturally derived anticancer agent, also inhibits Skp2 expression with concomitant CDKI upregulation. In line, curcumin suppressed cSCC cell growth through ROS-mediated apoptosis, while the use of N-acetyl cysteine (NAC) reversed curcumin-induced cell death. Curcumin treatment also sensitized cSCC cells to conventional anticancer drugs, such as cisplatin and doxorubicin. Altogether, these data suggest that Skp2 targeting restores the functioning of tumor suppressors, inhibits the expression of genes associated with cell proliferation and stemness, and sensitizes cancer cells to anticancer drugs. Thus, genetic, and pharmacological ablation of Skp2 can be an important strategy for attenuating cancer pathogenesis and associated complications in skin squamous cell carcinoma.
Collapse
Affiliation(s)
- Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Maha Al-Tamimi
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Rasheeda Anver
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Maha Victor Agha
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Gazala Anamangadan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow 226003, India
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar; Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; College of Medicine, Qatar University, Doha 2713, Qatar.
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar.
| |
Collapse
|
4
|
Li W, Jiang H, Zhang W, Sun Q, Zhang Q, Xu J, Huang J, Wan Y. Mechanisms of action of Sappan lignum for prostate cancer treatment: network pharmacology, molecular docking and experimental validation. Front Pharmacol 2024; 15:1407525. [PMID: 39318781 PMCID: PMC11420528 DOI: 10.3389/fphar.2024.1407525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the most common non-cutaneous malignancy in men globally. Sappan lignum, which exists in the heartwood of Caesalpinia sappan L., has antitumor effects; however, its exact mechanism of action remains unclear. This study elucidated the underlying mechanisms of Sappan lignum in PCa through network pharmacology approaches and molecular docking techniques. Moreover, the therapeutic effects of Sappan lignum on PCa were verified through in vitro experiments. METHODS The constituent ingredients of Sappan lignum were retrieved from the HERB database. Active plant-derived compounds of Sappan lignum were screened based on gastrointestinal absorption and gastric drug properties. Disease targets for PCa were screened using unpaired and paired case datasets from the Gene Expression Omnibus. Intersection targets were used for gene ontology and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis. Core targets were identified through topological analysis parameters and their clinical relevance was validated through The Cancer Genome Atlas database. The affinity between the phytochemicals of Sappan lignum and core proteins was verified using the molecular docking technique. Validation experiments confirmed the significant potential of Sappan lignum in treating PCa. RESULTS Twenty-one plant-derived compounds of Sappan lignum and 821 differentially expressed genes associated with PCa were collected. Among 32 intersection targets, 8 were screened according to topological parameters. KEGG analysis indicated that the antitumor effects of Sappan lignum on PCa were primarily associated with the p53 pathway. The molecular docking technique demonstrated a strong affinity between 3-deoxysappanchalcone (3-DSC) and core proteins, particularly cyclin B1 (CCNB1). CCNB1 expression correlated with clinicopathological features in patients with PCa. Experimental results revealed that 3-DSC exhibited anti-proliferative, anti-migratory, and pro-apoptotic effects on 22RV1 and DU145 cells while also causing G2/M phase cell cycle arrest, potentially through modulating the p53/p21/CDC2/CCNB1 pathway. CONCLUSION This research highlights the promising therapeutic potential of Sappan lignum in treating PCa, with a particular focus on targeting the p53 pathway.
Collapse
Affiliation(s)
- Wenna Li
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Acupuncture and Moxibustion in Cancer Care, Beijing University of Chinese Medicine, Beijing, China
| | - Honglin Jiang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weina Zhang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuyue Sun
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qiaoli Zhang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Acupuncture and Moxibustion in Cancer Care, Beijing University of Chinese Medicine, Beijing, China
| | - Jingnan Xu
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Acupuncture and Moxibustion in Cancer Care, Beijing University of Chinese Medicine, Beijing, China
| | - Jinchang Huang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Acupuncture and Moxibustion in Cancer Care, Beijing University of Chinese Medicine, Beijing, China
| | - Yuxiang Wan
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Acupuncture and Moxibustion in Cancer Care, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Hao Y, Gu C, Luo W, Shen J, Xie F, Zhao Y, Song X, Han Z, He J. The role of protein post-translational modifications in prostate cancer. PeerJ 2024; 12:e17768. [PMID: 39148683 PMCID: PMC11326433 DOI: 10.7717/peerj.17768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024] Open
Abstract
Involving addition of chemical groups or protein units to specific residues of the target protein, post-translational modifications (PTMs) alter the charge, hydrophobicity, and conformation of a protein, which in turn influences protein function, protein-protein interaction, and protein aggregation. These alterations, which include phosphorylation, glycosylation, ubiquitination, methylation, acetylation, lipidation, and lactylation, are significant biological events in the development of cancer, and play vital roles in numerous biological processes. The processes behind essential functions, the screening of clinical illness signs, and the identification of therapeutic targets all depend heavily on further research into the PTMs. This review outlines the influence of several PTM types on prostate cancer (PCa) diagnosis, therapy, and prognosis in an effort to shed fresh light on the molecular causes and progression of the disease.
Collapse
Affiliation(s)
- Yinghui Hao
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chenqiong Gu
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Shen
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangmei Xie
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Zhao
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyu Song
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zeping Han
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinhua He
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
- Rehabilitation Medicine Institute of Panyu District, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Wang J, Huang D, Nguyen TAT, Phan LM, Wei W, Rezaeian AH. CD74-AKT Axis Is a Potential Therapeutic Target in Triple-Negative Breast Cancer. BIOLOGY 2024; 13:481. [PMID: 39056676 PMCID: PMC11274071 DOI: 10.3390/biology13070481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/19/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Triple-negative breast cancer (TNBC) cells are often resistant to FAS (CD95)-mediated apoptosis, but the underlying molecular mechanism(s) is not fully understood yet. Notably, the expression of the type II transmembrane protein, CD74, is correlated with chemotherapy-resistant and more invasive forms of cancers via unknown mechanisms. Here, we analyzed gene expression pattern of cancer patients and/or patient-derived xenograft (PDX) models and found that mRNA and protein levels of CD74 are highly expressed in TNBC and correlated with cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT) properties. Mechanistically, we found that AKT activation is likely critical for maintaining CD74 expression and protein stability to favor its oncogenic functions. Physiologically, epidermal growth factor (EGF) along with CD74 could activate AKT signaling, likely through binding of phosphorylated AKT (S473) to CD74, whereas inhibition of AKT could impair stability of CD74. We also revealed that CD74 binds to FAS and interferes with the intrinsic signaling of FAS-mediated apoptosis. As such, selective targeting of the CD74/FAS complex using the AKT inhibitor along with the CD74-derived peptide could synergistically restore and activate FAS-mediated apoptosis. Therefore, our approach of mobilizing apoptosis pathways likely provides a rationale for TNBC treatment by targeting the CD74/FAS and CD74-AKT axes.
Collapse
Affiliation(s)
- Jingchao Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daoyuan Huang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Thu Anh Thai Nguyen
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Liem Minh Phan
- David Grant USAF Medical Center, Clinical Investigation Facility, 60th Medical Group, Travis Air Force Base, Fairfield, CA 94535, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
7
|
He YF, Liu YP, Liao JZ, Gan Y, Li X, Wang RR, Wang F, Zhou J, Zhou L. Xanthohumol Promotes Skp2 Ubiquitination Leading to the Inhibition of Glycolysis and Tumorigenesis in Ovarian Cancer. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:865-884. [PMID: 38790085 DOI: 10.1142/s0192415x24500356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Ovarian cancer is a common, highly lethal tumor. Herein, we reported that S-phase kinase-associated protein 2 (Skp2) is essential for the growth and aerobic glycolysis of ovarian cancer cells. Skp2 was upregulated in ovarian cancer tissues and associated with poor clinical outcomes. Using a customized natural product library screening, we found that xanthohumol inhibited aerobic glycolysis and cell viability of ovarian cancer cells. Xanthohumol facilitated the interaction between E3 ligase Cdh1 and Skp2 and promoted the Ub-K48-linked polyubiquitination of Skp2 and degradation. Cdh1 depletion reversed xanthohumol-induced Skp2 downregulation, enhancing HK2 expression and glycolysis in ovarian cancer cells. Finally, a xenograft tumor model was employed to examine the antitumor efficacy of xanthohumol in vivo. Collectively, we discovered that xanthohumol promotes the binding between Skp2 and Cdh1 to suppress the Skp2/AKT/HK2 signal pathway and exhibits potential antitumor activity for ovarian cancer cells.
Collapse
Affiliation(s)
- Yi-Fu He
- Department of Obstetrics and Gynecology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| | - Yi-Ping Liu
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| | - Jin-Zhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Rui-Rui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, P. R. China
| | - Fang Wang
- Tengzhou Central People's Hospital, Tengzhou 277500, P. R. China
| | - Jun Zhou
- Department of Medical Science Research Center, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| |
Collapse
|
8
|
Zhuang M, Li F, Liang H, Su Y, Cheng L, Lin B, Zhou J, Deng R, Chen L, Lyu P, Lu Z. Targeting RCC1 to block the human soft-tissue sarcoma by disrupting nucleo-cytoplasmic trafficking of Skp2. Cell Death Dis 2024; 15:241. [PMID: 38561375 PMCID: PMC10985091 DOI: 10.1038/s41419-024-06629-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Soft-tissue sarcomas (STS) emerges as formidable challenges in clinics due to the complex genetic heterogeneity, high rates of local recurrence and metastasis. Exploring specific targets and biomarkers would benefit the prognosis and treatment of STS. Here, we identified RCC1, a guanine-nucleotide exchange factor for Ran, as an oncogene and a potential intervention target in STS. Bioinformatics analysis indicated that RCC1 is highly expressed and correlated with poor prognosis in STS. Functional studies showed that RCC1 knockdown significantly inhibited the cell cycle transition, proliferation and migration of STS cells in vitro, and the growth of STS xenografts in mice. Mechanistically, we identified Skp2 as a downstream target of RCC1 in STS. Loss of RCC1 substantially diminished Skp2 abundance by compromising its protein stability, resulting in the upregulation of p27Kip1 and G1/S transition arrest. Specifically, RCC1 might facilitate the nucleo-cytoplasmic trafficking of Skp2 via direct interaction. As a result, the cytoplasmic retention of Skp2 would further protect it from ubiquitination and degradation. Notably, recovery of Skp2 expression largely reversed the phenotypes induced by RCC1 knockdown in STS cells. Collectively, this study unveils a novel RCC1-Skp2-p27Kip1 axis in STS oncogenesis, which holds promise for improving prognosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mingzhi Zhuang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Fengyue Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Hong Liang
- College of Geography and Oceanography, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, 350108, P. R. China
| | - Yongfu Su
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Lei Cheng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Bingkai Lin
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Jun Zhou
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Runzhi Deng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Linying Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, P. R. China
| | - Peng Lyu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China.
| | - Zhonglei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China.
| |
Collapse
|
9
|
Wang J, Wang Z, Inuzuka H, Wei W, Liu J. PRMT1 methylates METTL14 to modulate its oncogenic function. Neoplasia 2023; 42:100912. [PMID: 37269817 PMCID: PMC10248872 DOI: 10.1016/j.neo.2023.100912] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/05/2023]
Abstract
N6-methyladenosine (m6A), the most abundant mRNA modification in mammalian cells, is responsible for mRNA stability and alternative splicing. The METTL3-METTL14-WTAP complex is the only methyltransferase for the m6A modification. Thus, regulation of its enzymatic activity is critical for the homeostasis of mRNA m6A levels in cells. However, relatively little is known about the upstream regulation of the METTL3-METTL14-WTAP complex, especially at the post-translational modification level. The C-terminal RGG repeats of METTL14 are critical for RNA binding. Therefore, modifications on these residues may play a regulatory role in its function. Arginine methylation is a post-translational modification catalyzed by protein arginine methyltransferases (PRMTs), among which PRMT1 preferentially methylates protein substrates with an arginine/glycine-rich motif. In addition, PRMT1 functions as a key regulator of mRNA alternative splicing, which is associated with m6A modification. To this end, we report that PRMT1 promotes the asymmetric methylation of two major arginine residues at the C-terminus of METTL14, and the reader protein SPF30 recognizes this modification. Functionally, PRMT1-mediated arginine methylation on METTL14 is likely essential for its function in catalyzing the m6A modification. Moreover, arginine methylation of METTL14 promotes cell proliferation that is antagonized by the PRMT1 inhibitor MS023. These results indicate that PRMT1 likely regulates m6A modification and promotes tumorigenesis through arginine methylation at the C-terminus of METTL14.
Collapse
Affiliation(s)
- Jingchao Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zhen Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jing Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|