1
|
Wan D, He L, Guo C, Zhong Z, Yan X, Cao J, Xu Q, Zhang H, Duan B. m6A-related lncRNAs predict prognosis and indicate cell cycle in gastric cancer. Front Genet 2023; 14:1140218. [PMID: 37408779 PMCID: PMC10319253 DOI: 10.3389/fgene.2023.1140218] [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: 01/29/2023] [Accepted: 04/11/2023] [Indexed: 07/07/2023] Open
Abstract
Background: N6-methyladenosine (m6A) modification is a common epigenetic methylation modification of RNA, which plays an important role in gastric carcinogenesis and progression by regulating long non-coding RNA (lncRNA). This study is aimed to investigate the potential prognostic signatures of m6A -related lncRNAs in STAD. Methods: The m6A-related lncRNAs with the most significant impact on gastric cancer prognosis in the TCGA database were identified by bioinformatics and machine learning methods. The m6A-related lncRNA prognostic model (m6A-LPS) and nomogram was constructed by Cox regression analysis with the minimum absolute contraction and selection operator (LASSO) algorithm. The functional enrichment analysis of m6A-related lncRNAs was also investigated. The miRTarBase, miRDB and TargetScan databases were utilized to establish a prognosis-related network of competing endogenous RNA (ceRNA) by bioinformatics methods. The correlation of AL391152.1 expressions and cell cycle were experimentally testified by qRT-PCR and flow cytometry. Results: In total, 697 lncRNAs that were identified as m6A-related lncRNAs in GC samples. The survival analysis showed that 18 lncRNAs demonstrated prognostic values. A risk model with 11 lncRNAs was established by Lasso Cox regression, and can predict the prognosis of GC patients. Cox regression analysis and ROC curve indicated that this lncRNA prediction model was an independent risk factor for survival rates. Functional enrichment analysis and ceRNA network revealed that the nomogram was notably associated with cell cycle. qRT-PCR and flow cytometry revealed that downregulation of GC m6A-related lncRNA AL391152.1 could decrease cyclins expression in SGC7901 cells. Conclusion: A m6A-related lncRNAs prognostic model was established in this study, which can be applied to predict prognosis and cell cycle in gastric cancer.
Collapse
Affiliation(s)
- Dong Wan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lingnan He
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Guo
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zishao Zhong
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xiaohan Yan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Cao
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qinwei Xu
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haibin Zhang
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bensong Duan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Breviscapine Participates in the Progression of Prostate Cancer by Inhibiting ZFP91 Expression through Upregulation of MicroRNA-129-5p. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:1511607. [PMID: 34925523 PMCID: PMC8674053 DOI: 10.1155/2021/1511607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 11/23/2022]
Abstract
Objective To investigate the effect of breviscapine (BVP) on the development of prostate cancer and its molecular mechanism. Materials and Methods After treatment with breviscapine and microRNA-129-5p, MTT (3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide) and cell counting kit-8 (CCK-8) tests were performed to examine the proliferation rate of cells, while Transwell was used to analyze cell migration ability; at the same time, quantitative real-time polymerase chain reaction (qRT-PCR) was applied to detect the expression of microRNA-129-5p and ZFP91 in prostate cancer cells. In addition, the binding of microRNA-129-5p and ZFP91 was confirmed by dual-luciferase reporting assay; meanwhile, cell reverse experiment verified that breviscapine can regulate ZFP91 via upregulating microRNA-129-5p. Results The results of MTT, CCK-8, and Transwell experiments demonstrated that breviscapine inhibited the proliferation as well as the migration capacities of PC cells; meanwhile, it upregulated the level of microRNA-129-5p in PC cells while downregulated that of ZFP91. Furthermore, dual-luciferase reporter gene assay verified that ZFP91 was a potential target of microRNA-129-5p. Finally, cell reverse experiment confirmed that breviscapine downregulated ZFP91 expression by upregulating microRNA-129-5p, while downregulation of microRNA-129-5p partially reversed the inhibitory effect of breviscapine on cell proliferation ability. Conclusions Breviscapine may inhibit the expression of ZFP91 through upregulating microRNA-129-5p and thus participating in the progression of PC.
Collapse
|
3
|
Liu Y, Duan C, Zhang C. E3 Ubiquitin Ligase in Anticancer Drugdsla Resistance: Recent Advances and Future Potential. Front Pharmacol 2021; 12:645864. [PMID: 33935743 PMCID: PMC8082683 DOI: 10.3389/fphar.2021.645864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/24/2021] [Indexed: 12/31/2022] Open
Abstract
Drug therapy is the primary treatment for patients with advanced cancer. The use of anticancer drugs will inevitably lead to drug resistance, which manifests as tumor recurrence. Overcoming chemoresistance may enable cancer patients to have better therapeutic effects. However, the mechanisms underlying drug resistance are poorly understood. E3 ubiquitin ligases (E3s) are a large class of proteins, and there are over 800 putative functional E3s. E3s play a crucial role in substrate recognition and catalyze the final step of ubiquitin transfer to specific substrate proteins. The diversity of the set of substrates contributes to the diverse functions of E3s, indicating that E3s could be desirable drug targets. The E3s MDM2, FBWX7, and SKP2 have been well studied and have shown a relationship with drug resistance. Strategies targeting E3s to combat drug resistance include interfering with their activators, degrading the E3s themselves and influencing the interaction between E3s and their substrates. Research on E3s has led to the discovery of possible therapeutic methods to overcome the challenging clinical situation imposed by drug resistance. In this article, we summarize the role of E3s in cancer drug resistance from the perspective of drug class.
Collapse
Affiliation(s)
- Yuanqi Liu
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| |
Collapse
|
4
|
Wang A, Ding L, Wu Z, Ding R, Teng XL, Wang F, Hu Z, Chen L, Yu X, Zou Q. ZFP91 is required for the maintenance of regulatory T cell homeostasis and function. J Exp Med 2021; 218:211637. [PMID: 33355624 PMCID: PMC7769166 DOI: 10.1084/jem.20201217] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/30/2020] [Accepted: 11/18/2020] [Indexed: 02/04/2023] Open
Abstract
Autophagy programs the metabolic and functional fitness of regulatory T (T reg) cells to establish immune tolerance, yet the mechanisms governing autophagy initiation in T reg cells remain unclear. Here, we show that the E3 ubiquitin ligase ZFP91 facilitates autophagy activation to sustain T reg cell metabolic programming and functional integrity. T reg cell-specific deletion of Zfp91 caused T reg cell dysfunction and exacerbated colonic inflammation and inflammation-driven colon carcinogenesis. TCR-triggered autophagy induction largely relied on T reg cell-derived ZFP91 to restrict hyperglycolysis, which is required for the maintenance of T reg cell homeostasis. Mechanistically, ZFP91 rapidly translocated from the nucleus to the cytoplasm in response to TCR stimulation and then mediated BECN1 ubiquitination to promote BECN1-PIK3C3 complex formation. Therefore, our results highlight a ZFP91-dependent mechanism promoting TCR-initiated autophagosome maturation to maintain T reg cell homeostasis and function.
Collapse
Affiliation(s)
- Aiting Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Ding
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongqiu Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Ding
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Lu Teng
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feixiang Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhilin Hu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Yu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Zou
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Hanafi M, Chen X, Neamati N. Discovery of a Napabucasin PROTAC as an Effective Degrader of the E3 Ligase ZFP91. J Med Chem 2021; 64:1626-1648. [PMID: 33506674 DOI: 10.1021/acs.jmedchem.0c01897] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Napabucasin, undergoing multiple clinical trials, was reported to inhibit the signal transducer and transcription factor 3 (STAT3). To better elucidate its mechanism of action, we designed a napabucasin-based proteolysis targeting chimera (PROTAC), XD2-149 that resulted in inhibition of STAT3 signaling in pancreatic cancer cell lines without inducing proteasome-dependent degradation of STAT3. Proteomics analysis of XD2-149 revealed the downregulation of the E3 ubiquitin-protein ligase ZFP91. XD2-149 degrades ZFP91 with DC50 values in the nanomolar range. The cytotoxicity of XD2-149 was significantly, but not fully, reduced with ZFP91 knockdown providing evidence for its multi-targeted mechanism of action. The NQO1 inhibitor, dicoumarol, rescued the cytotoxicity of XD2-149 but not ZFP91 degradation, suggesting that the NQO1-induced cell death is independent of ZFP91. ZFP91 plays a role in tumorigenesis and is involved in multiple oncogenic pathways including NF-κB and HIF-1α.
Collapse
Affiliation(s)
- Maha Hanafi
- Department of Medicinal Chemistry, College of Pharmacy and the Rogel Cancer Center, North Campus Research Complex, 1600 Huron Parkway, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Xinde Chen
- Department of Medicinal Chemistry, College of Pharmacy and the Rogel Cancer Center, North Campus Research Complex, 1600 Huron Parkway, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy and the Rogel Cancer Center, North Campus Research Complex, 1600 Huron Parkway, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
6
|
Cui S, Lei Z, Guan T, Fan L, Li Y, Geng X, Fu D, Jiang H, Xu S. Targeting USP1-dependent KDM4A protein stability as a potential prostate cancer therapy. Cancer Sci 2020; 111:1567-1581. [PMID: 32133742 PMCID: PMC7226285 DOI: 10.1111/cas.14375] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/08/2020] [Accepted: 02/25/2020] [Indexed: 01/10/2023] Open
Abstract
The histone demethylase lysine-specific demethylase 4A (KDM4A) is reported to be overexpressed and plays a vital in multiple cancers through controlling gene expression by epigenetic regulation of H3K9 or H3K36 methylation marks. However, the biological role and mechanism of KDM4A in prostate cancer (PC) remain unclear. Herein, we reported KDM4A expression was upregulation in phosphatase and tensin homolog knockout mouse prostate tissue. Depletion of KDM4A in PC cells inhibited their proliferation and survival in vivo and vitro. Further studies reveal that USP1 is a deubiquitinase that regulates KDM4A K48-linked deubiquitin and stability. Interestingly, we found c-Myc was a key downstream effector of the USP1-KDM4A/androgen receptor axis in driving PC cell proliferation. Notably, upregulation of KDM4A expression with high USP1 expression was observed in most prostate tumors and inhibition of USP1 promotes PC cells response to therapeutic agent enzalutamide. Our studies propose USP1 could be an anticancer therapeutic target in PC.
Collapse
Affiliation(s)
- Shu‐Zhong Cui
- Department of Abdominal SurgeryAffiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Zi‐Ying Lei
- Department of Abdominal SurgeryAffiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Tian‐Pei Guan
- Department of Abdominal SurgeryAffiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Ling‐Ling Fan
- Department of BiochemistryMarlene and Stewart Greenebaum Cancer CenterUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - You‐Qiang Li
- Department of BiochemistryMarlene and Stewart Greenebaum Cancer CenterUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Xin‐Yan Geng
- Department of BiochemistryMarlene and Stewart Greenebaum Cancer CenterUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - De‐Xue Fu
- Department of SurgeryMarlene and Stewart Greenebaum Cancer CenterUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Hao‐Wu Jiang
- Department of AnesthesiologyCenter for the Study of ItchWashington University School of MedicineSt. LouisMOUSA
| | - Song‐Hui Xu
- Department of Abdominal SurgeryAffiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhouChina
- Department of BiochemistryMarlene and Stewart Greenebaum Cancer CenterUniversity of Maryland School of MedicineBaltimoreMDUSA
| |
Collapse
|