1
|
Feng T, Wang P, Zhang X. Skp2: A critical molecule for ubiquitination and its role in cancer. Life Sci 2024; 338:122409. [PMID: 38184273 DOI: 10.1016/j.lfs.2023.122409] [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: 10/09/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
The ubiquitin-proteasome system (UPS) is a multi-step process that serves as the primary pathway for protein degradation within cells. UPS activity also plays a crucial role in regulating various life processes, including the cell cycle, signal transduction, DNA repair, and others. The F-box protein Skp2, a crucial member of the UPS, plays a central role in the development of various diseases. Skp2 controls cancer cell growth and drug resistance by ubiquitinating modifications to a variety of proteins. This review emphasizes the multifaceted role of Skp2 in a wide range of cancers and the mechanisms involved, highlighting the potential of Skp2 as a therapeutic target in cancer. Additionally, we describe the impactful influence exerted by Skp2 in various other diseases beyond cancer.
Collapse
Affiliation(s)
- Tianyang Feng
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China
| | - Ping Wang
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China
| | - Xiling Zhang
- The Fourth Affiliated Hospital of China Medical University, Department of Urology, Shenyang 110032, China; Liaoning Provincial Key Laboratory of Basic Research for Bladder Diseases, Shenyang 110000, China.
| |
Collapse
|
2
|
Zhang C, Pan G, Qin JJ. Role of F-box proteins in human upper gastrointestinal tumors. Biochim Biophys Acta Rev Cancer 2024; 1879:189035. [PMID: 38049014 DOI: 10.1016/j.bbcan.2023.189035] [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: 08/23/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/06/2023]
Abstract
Protein ubiquitination and degradation is an essential physiological process in almost all organisms. As the key participants in this process, the E3 ubiquitin ligases have been widely studied and recognized. F-box proteins, a crucial component of E3 ubiquitin ligases that regulates diverse biological functions, including cell differentiation, proliferation, migration, and apoptosis by facilitating the degradation of substrate proteins. Currently, there is an increasing focus on studying the role of F-box proteins in cancer. In this review, we present a comprehensive overview of the significant contributions of F-box proteins to the development of upper gastrointestinal tumors, highlighting their dual roles as both carcinogens and tumor suppressors. We delve into the molecular mechanisms underlying the involvement of F-box proteins in upper gastrointestinal tumors, exploring their interactions with specific substrates and their cross-talks with other key signaling pathways. Furthermore, we discuss the implications of F-box proteins in radiotherapy resistance in the upper gastrointestinal tract, emphasizing their potential as clinical therapeutic and prognostic targets. Overall, this review provides an up-to-date understanding of the intricate involvement of F-box proteins in human upper gastrointestinal tumors, offering valuable insights for the identification of prognostic markers and the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Che Zhang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guangzhao Pan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jiang-Jiang Qin
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
3
|
Qiao C, Huang F, He J, Wu Q, Zheng Z, Zhang T, Miao Y, Yuan Y, Chen X, Du Q, Xu Y, Wu D, Yu Z, Zheng H. Ceftazidime reduces cellular Skp2 to promote type-I interferon activity. Immunology 2023; 170:527-539. [PMID: 37641430 DOI: 10.1111/imm.13687] [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: 12/17/2022] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Skp2 plays multiple roles in malignant tumours. Here, we revealed that Skp2 negatively regulates type-I interferon (IFN-I)-mediated antiviral activity. We first noticed that Skp2 can promote virus infection in cells. Further studies demonstrated that Skp2 interacts with IFN-I receptor 2 (IFNAR2) and promotes K48-linked polyubiquitination of IFNAR2, which accelerates the degradation of IFNAR2 proteins. Skp2-mediated downregulation of IFNAR2 levels inhibits IFN-I signalling and IFN-I-induced antiviral activity. In addition, we uncovered for the first time that the antibiotic ceftazidime can act as a repressor of Skp2. Ceftazidime reduces cellular Skp2 levels, thus enhancing IFNAR2 stability and IFN-I antiviral activity. This study reveals a new role of Skp2 in regulating IFN-I signalling and IFN-I antiviral activity and reports the antibiotic ceftazidime as a potential repressor of Skp2.
Collapse
Affiliation(s)
- Caixia Qiao
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Fan Huang
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
- The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jiuyi He
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Qiuyu Wu
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Zhijin Zheng
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Tingting Zhang
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Ying Miao
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yukang Yuan
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Xiangjie Chen
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Qian Du
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Zhengyuan Yu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hui Zheng
- Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Yang H, Ai H, Zhang J, Ma J, Liu K, Li Z. UPS: Opportunities and challenges for gastric cancer treatment. Front Oncol 2023; 13:1140452. [PMID: 37077823 PMCID: PMC10106573 DOI: 10.3389/fonc.2023.1140452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Gastric cancer remains the fourth most frequently diagnosed malignancy and the fifth leading cause of cancer-related mortality worldwide owning to the lack of efficient drugs and targets for therapy. Accumulating evidence indicates that UPS, which consists of E1, E2, and E3 enzymes and proteasome, plays an important role in the GC tumorigenesis. The imbalance of UPS impairs the protein homeostasis network during development of GC. Therefore, modulating these enzymes and proteasome may be a promising strategy for GC target therapy. Besides, PROTAC, a strategy using UPS to degrade the target protein, is an emerging tool for drug development. Thus far, more and more PROTAC drugs enter clinical trials for cancer therapy. Here, we will analyze the abnormal expression enzymes in UPS and summarize the E3 enzymes which can be developed in PROTAC so that it can contribute to the development of UPS modulator and PROTAC technology for GC therapy.
Collapse
Affiliation(s)
- Hang Yang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Huihan Ai
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jialin Zhang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jie Ma
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US Hormel (Henan) Cancer Institute, Zhengzhou, Henan, China
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Zhi Li, ; Kangdong Liu,
| | - Zhi Li
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Zhi Li, ; Kangdong Liu,
| |
Collapse
|
5
|
Tanaka N, Sakamoto T. Mint3 as a Potential Target for Cooling Down HIF-1α-Mediated Inflammation and Cancer Aggressiveness. Biomedicines 2023; 11:biomedicines11020549. [PMID: 36831085 PMCID: PMC9953510 DOI: 10.3390/biomedicines11020549] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 02/16/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that plays a crucial role in cells adapting to a low-oxygen environment by facilitating a switch from oxygen-dependent ATP production to glycolysis. Mediated by membrane type-1 matrix metalloproteinase (MT1-MMP) expression, Munc-18-1 interacting protein 3 (Mint3) binds to the factor inhibiting HIF-1 (FIH-1) and inhibits its suppressive effect, leading to HIF-1α activation. Defects in Mint3 generally lead to improved acute inflammation, which is regulated by HIF-1α and subsequent glycolysis, as well as the suppression of the proliferation and metastasis of cancer cells directly through its expression in cancer cells and indirectly through its expression in macrophages or fibroblasts associated with cancer. Mint3 in inflammatory monocytes enhances the chemotaxis into metastatic sites and the production of vascular endothelial growth factors, which leads to the expression of E-selectin at the metastatic sites and the extravasation of cancer cells. Fibroblasts express L1 cell adhesion molecules in a Mint3-dependent manner and enhance integrin-mediated cancer progression. In pancreatic cancer cells, Mint3 directly promotes cancer progression. Naphthofluorescein, a Mint3 inhibitor, can disrupt the interaction between FIH-1 and Mint3 and potently suppress Mint3-mediated inflammation, cancer progression, and metastasis without causing marked adverse effects. In this review, we will introduce the potential of Mint3 as a therapeutic target for inflammatory diseases and cancers.
Collapse
|
6
|
Jing J, Rui L, Junyuan S, Jinfeng Y, Zhihao H, Weiguo L, Zhenyu J. Small-molecule compounds inhibiting S-phase kinase-associated protein 2: A review. Front Pharmacol 2023; 14:1122008. [PMID: 37089937 PMCID: PMC10113621 DOI: 10.3389/fphar.2023.1122008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
S-phase kinase-associated protein 2 (Skp2) is a substrate-specific adaptor in Skp1-CUL1-ROC1-F-box E3 ubiquitin ligases and widely regarded as an oncogene. Therefore, Skp2 has remained as an active anticancer research topic since its discovery. Accordingly, the structure of Skp2 has been solved and numerous Skp2 inhibiting compounds have been identified. In this review, we would describe the structural features of Skp2, introduce the ubiquitination function of SCFSkp2, and summarize the diverse natural and synthetic Skp2 inhibiting compounds reported to date. The IC50 data of the Skp2 inhibitors or inhibiting compounds in various kinds of tumors at cellular levels implied that the cancer type, stage and pathological mechanisms should be taken into consideration when selecting Skp2-inhibiting compound for cancer treatment.
Collapse
Affiliation(s)
- Jia Jing
- Schools of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Li Rui
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Sun Junyuan
- Schools of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Yang Jinfeng
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Hong Zhihao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Lu Weiguo
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Women′s Reproductive Health Research of Zhejiang Province, Hangzhou, Zhejiang Province, China
- *Correspondence: Lu Weiguo, ; Jia Zhenyu,
| | - Jia Zhenyu
- Institute of Occupation Diseases, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
- *Correspondence: Lu Weiguo, ; Jia Zhenyu,
| |
Collapse
|
7
|
Liu J, Chen T, Li S, Liu W, Wang P, Shang G. Targeting matrix metalloproteinases by E3 ubiquitin ligases as a way to regulate the tumor microenvironment for cancer therapy. Semin Cancer Biol 2022; 86:259-268. [PMID: 35724822 DOI: 10.1016/j.semcancer.2022.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/27/2022] [Accepted: 06/13/2022] [Indexed: 10/31/2022]
Abstract
The tumor microenvironment (TME) plays an important role in neoplastic development. Matrix metalloproteinases (MMPs) are critically involved in tumorigenesis by modulation of the TME and degradation of the extracellular matrix (ECM) in a large variety of malignancies. Evidence has revealed that dysregulated MMPs can lead to ECM damage, the promotion of cell migration and tumor metastasis. The expression and activities of MMPs can be tightly regulated by TIMPs, multiple signaling pathways and noncoding RNAs. MMPs are also finely controlled by E3 ubiquitin ligases. The current review focuses on the molecular mechanism by which MMPs are governed by E3 ubiquitin ligases in carcinogenesis. Due to the essential role of MMPs in oncogenesis, they have been considered the attractive targets for antitumor treatment. Several strategies that target MMPs have been discovered, including the use of small-molecule inhibitors, peptides, inhibitory antibodies, natural compounds with anti-MMP activity, and RNAi therapeutics. However, these molecules have multiple disadvantages, such as poor solubility, severe side-effects and low oral bioavailability. Therefore, it is necessary to discover the novel inhibitors that suppress MMPs for cancer therapy. Here, we discuss the therapeutic potential of targeting E3 ubiquitin ligases to inhibit MMPs. We hope this review will stimulate the discovery of novel therapeutics for the MMP-targeted treatment of a variety of human cancers.
Collapse
Affiliation(s)
- Jinxin Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Shizhe Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Wenjun Liu
- Department of Research and Development, Beijing Zhongwei Research Center of Biological and Translational Medicine, Beijing 100161, China
| | - Peter Wang
- Department of Research and Development, Beijing Zhongwei Research Center of Biological and Translational Medicine, Beijing 100161, China; Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Anhui 233030, China.
| | - Guanning Shang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
8
|
Wu T, Gu X, Cui H. Emerging Roles of SKP2 in Cancer Drug Resistance. Cells 2021; 10:cells10051147. [PMID: 34068643 PMCID: PMC8150781 DOI: 10.3390/cells10051147] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
More than half of all cancer patients receive chemotherapy, however, some of them easily acquire drug resistance. Resistance to chemotherapy has become a massive obstacle to achieve high rates of pathological complete response during cancer therapy. S-phase kinase-associated protein 2 (Skp2), as an E3 ligase, was found to be highly correlated with drug resistance and poor prognosis. In this review, we summarize the mechanisms that Skp2 confers to drug resistance, including the Akt-Skp2 feedback loop, Skp2-p27 pathway, cell cycle and mitosis regulation, EMT (epithelial-mesenchymal transition) property, enhanced DNA damage response and repair, etc. We also addressed novel molecules that either inhibit Skp2 expression or target Skp2-centered interactions, which might have vast potential for application in clinics and benefit cancer patients in the future.
Collapse
Affiliation(s)
- Ting Wu
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China;
| | - Xinsheng Gu
- Department of Pharmacology, College of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, China;
| | - Hongmei Cui
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China;
- Correspondence:
| |
Collapse
|
9
|
Wang H, Lu Y, Wang M, Wu Y, Wang X, Li Y. Roles of E3 ubiquitin ligases in gastric cancer carcinogenesis and their effects on cisplatin resistance. J Mol Med (Berl) 2021; 99:193-212. [PMID: 33392633 DOI: 10.1007/s00109-020-02015-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022]
Abstract
Although gastric cancer (GC) is one of the most common cancers with high incidence and mortality rates, its pathogenesis is still not elucidated. GC carcinogenesis is complicated and involved in the activation of oncoproteins and inactivation of tumor suppressors. The ubiquitin-proteasome system (UPS) is crucial for protein degradation and regulation of physiological and pathological processes. E3 ubiquitin ligases are pivotal enzymes in UPS, containing various subfamily proteins. Previous studies report that some E3 ligases, including SKP2, CUL1, and MDM2, act as oncoproteins in GC carcinogenesis. On the other hand, FBXW7, FBXL5, FBXO31, RNF43, and RNF180 exert as tumor suppressors in GC carcinogenesis. Moreover, E3 ligases modulate cell growth, cell apoptosis, and cell cycle; thus, it is complicated to confer cisplatin resistance/sensitivity in GC cells. The intrinsic and acquired cisplatin resistance limits its clinical application against GC. In this review, we explore oncogenic and tumor suppressive roles of E3 ligases in GC carcinogenesis and focus on the effects of E3 ligases on cisplatin resistance in GC cells, which will provide novel therapeutic targets for GC therapy, especially for cisplatin-resistant patients.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Mingliang Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Youliang Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaodong Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
10
|
Shan P, Tang B, Xie S, Zhang Z, Fan J, Wei Z, Song C. NDV-D90 inhibits 17β-estradiol-mediated resistance to apoptosis by differentially modulating classic and nonclassic estrogen receptors in breast cancer cells. J Cell Biochem 2020; 122:3-15. [PMID: 32985706 DOI: 10.1002/jcb.28118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/29/2018] [Indexed: 01/12/2023]
Abstract
Newcastle disease virus (NDV) is endowed with the oncolytic ability to kill tumor cells, while rarely causing side effects in normal cells. Both estrogen receptor α (ERα) and the G protein estrogen receptor (GPER) modulate multiple biological activities in response to estrogen, including apoptosis in breast cancer (BC) cells. Here, we investigated whether NDV-D90, a novel strain isolated from natural sources in China, promoted apoptosis by modulating the expression of ERα or the GPER in BC cells exposed to 17β-estradiol (E2). We found that NDV-D90 significantly killed the tumor cell lines MCF-7 and BT549 in a time- and dose-dependent manner. We also found that NDV-D90 exerted its effects on the two cell lines mainly by inducing apoptosis but not necrosis. NDV-D90 induced apoptosis via the intrinsic and extrinsic signaling pathways in MCF-7 cells (ER-positive cells) during E2 exposure not only by disrupting the E2/ERα axis and enhancing GPER expression but also by modulating the expression of several apoptosis-related proteins through ERα-and GPER-independent processes. NDV-D90 promoted apoptosis via the intrinsic signaling pathway in BT549 cells (ER-negative cells), possibly by impairing E2-mediated GPER expression. Furthermore, NDV-D90 exerted its antitumor effects in vivo by inducing apoptosis. Overall, these results demonstrated that NDV-D90 promotes apoptosis by differentially modulating the expression of ERα and the GPER in ER-positive and negative BC cells exposed to estrogen, respectively, and can be utilized as an effective approach to treating BC.
Collapse
Affiliation(s)
- Peng Shan
- Department of General Surgery, The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Tang
- Department of General Surgery, The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanshan Xie
- Department of Thyroid Gland and Breast Surgery, The Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Hubei, China
| | - Zengling Zhang
- Department of General Surgery, Central Hospital of Pukou District, Nanjing, China
| | - Jiehou Fan
- Department of Breast Surgery, The Second People's Hospital of Dezhou, Dezhou, China
| | - Zheng Wei
- Department of General Surgery, The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chun Song
- The Key Laboratory of Cell Transplantation of Ministry of Health and Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Asmamaw MD, Liu Y, Zheng YC, Shi XJ, Liu HM. Skp2 in the ubiquitin-proteasome system: A comprehensive review. Med Res Rev 2020; 40:1920-1949. [PMID: 32391596 DOI: 10.1002/med.21675] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/26/2020] [Accepted: 04/27/2020] [Indexed: 12/19/2022]
Abstract
The ubiquitin-proteasome system (UPS) is a complex process that regulates protein stability and activity by the sequential actions of E1, E2 and E3 enzymes to influence diverse aspects of eukaryotic cells. However, due to the diversity of proteins in cells, substrate selection is a highly critical part of the process. As a key player in UPS, E3 ubiquitin ligases recruit substrates for ubiquitination specifically. Among them, RING E3 ubiquitin ligases which are the most abundant E3 ubiquitin ligases contribute to diverse cellular processes. The multisubunit cullin-RING ligases (CRLs) are the largest family of RING E3 ubiquitin ligases with tremendous plasticity in substrate specificity and regulate a vast array of cellular functions. The F-box protein Skp2 is a component of CRL1 (the prototype of CRLs) which is expressed in many tissues and participates in multiple cellular functions such as cell proliferation, metabolism, and tumorigenesis by contributing to the ubiquitination and subsequent degradation of several specific tumor suppressors. Most importantly, Skp2 plays a pivotal role in a plethora of cancer-associated signaling pathways. It enhances cell growth, accelerates cell cycle progression, promotes migration and invasion, and inhibits cell apoptosis among others. Hence, targeting Skp2 may represent a novel and attractive strategy for the treatment of different human cancers overexpressing this oncogene. In this review article, we summarized the known roles of Skp2 both in health and disease states in relation to the UPS.
Collapse
Affiliation(s)
- Moges Dessale Asmamaw
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Ying Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Xiao-Jing Shi
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Pine needle hexane extract promote cell cycle arrest and premature senescence via p27 KIP1 upregulation gastric cancer cells. Food Sci Biotechnol 2020; 29:845-853. [PMID: 32523794 DOI: 10.1007/s10068-019-00730-5] [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] [Received: 07/23/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 01/06/2023] Open
Abstract
Pinus densiflora sieb. et zucc.(pine needle) is a traditional medicine used in several East Asian countries. However, the efficacy of pine needle has rarely been reported. In this study showed that the anti-proliferative effects and the mechanisms of hexane layer of pine needle MeOH extract (PNH) on gastric cancer cells. At first, PNH inhibited the proliferation of gastric cancer cells in a dose-dependent manner. Moreover, PNH treatment induced G1 phase cell cycle arrest through the increased p27KIP1 expression and decreased cyclin dependent kinase (CDKs) activity. Furthermore, PNH treatment induced premature senescence without oncogenic stress, through the expression of p27KIP1 and Skp2. Taken together, these results showed that PNH inhibited gastric cancer cell proliferation through the induction of G1-cell cycle arrest and premature senescence via induced p27KIP1 expression, as controlled by Skp2 reduction. Also, PNH could be a candidate for anti-gastric cancer treatment and may be useful in the development of anti-gastric cancer drugs.
Collapse
|
13
|
Jingyue S, Xiao W, Juanmin Z, Wei L, Daoming L, Hong X. TFAP2E methylation promotes 5‑fluorouracil resistance via exosomal miR‑106a‑5p and miR‑421 in gastric cancer MGC‑803 cells. Mol Med Rep 2019; 20:323-331. [PMID: 31115533 PMCID: PMC6579997 DOI: 10.3892/mmr.2019.10237] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/18/2019] [Indexed: 12/24/2022] Open
Abstract
Hypermethylation of transcription factor activating enhancer-binding protein 2e (TFAP2E) has been reported to be associated with chemoresistance to 5-fluorouracil (5-FU) in gastric cancer (GC). In the present study, the molecular mechanism governing this chemoresistance was investigated. Drug-resistant human GC MGC-803/5-FU cells were established and TFAP2E expression and methylation levels were assessed. Autocrine exosomes from GC culture medium were isolated and characterized. MicroRNA (miRNA) microarray analysis was used to determine the miRNA expression profile of GC cell-derived exosomes. Exosomes collected from MGC-803/5-FU cells were co-cultured with control cells, and 5-Aza-2′-deoxycytidine (5Aza) was added into MGC-803/5-FU cells to investigate the relationship between TFAP2E, exosomes and chemosensitivity. In the present study, it was demonstrated that hypermethylation of TFAP2E resulted in its reduced expression and 5-FU chemoresistance in GC cells. miRNAs miR-106a-5p and miR-421 were highly expressed and regulated the chemoresistance induced by TFAP2E methylation. Target gene prediction using miRBase, TargetScan and PicTar revealed that E2F1, MTOR and STAT3 may be TFAP2E target genes in GC. Collectively, our data support an important role of exosomes and exosomal miRNAs in TFAP2E methylation-induced chemoresistance to 5-FU in GC. These results highlight their potential for miRNA-based therapeutics.
Collapse
Affiliation(s)
- Sun Jingyue
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wang Xiao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Zha Juanmin
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Li Wei
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Li Daoming
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xu Hong
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
14
|
Identification and structural characterization of deleterious non-synonymous single nucleotide polymorphisms in the human SKP2 gene. Comput Biol Chem 2019; 79:127-136. [PMID: 30802828 DOI: 10.1016/j.compbiolchem.2019.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 01/27/2019] [Accepted: 02/13/2019] [Indexed: 12/17/2022]
Abstract
In SCF (Skp, Cullin, F-box) ubiquitin-protein ligase complexes, S-phase kinase 2 (SKP2) is one of the major players of F-box family, that is responsible for the degradation of several important cell regulators and tumor suppressor proteins. Despite of having significant evidence for the role of SKP2 on tumorgenesis, there is a lack of available data regarding the effect of non-synonymous polymorphisms. In this communication, the structural and functional consequences of non-synonymous single nucleotide polymorphisms (nsSNPs) of SKP2 have been reported by employing various computational approaches and molecular dynamics simulation. Initially, several computational tools like SIFT, PolyPhen-2, PredictSNP, I-Mutant 2.0 and ConSurf have been implicated in this study to explore the damaging SNPs. In total of 172 nsSNPs, 5 nsSNPs were identified as deleterious and 3 of them were predicted to be decreased the stability of protein. Guided from ConSurf analysis, P101L (rs761253702) and Y346C (rs755010517) were categorized as the highly conserved and functional disrupting mutations. Therefore, these mutations were subjected to three dimensional model building and molecular dynamics simulation study for the detailed structural consequences upon the mutations. The study revealed that P101L and Y346C mutations increased the flexibility and changed the structural dynamics. As both these mutations are located in the most functional regions of SKP2 protein, these computational insights might be helpful to consider these nsSNPs for wet-lab confirmatory analysis as well as in rationalizing future population based studies and structure based drug design against SKP2.
Collapse
|
15
|
Shang HS, Lu HF, Lee CH, Chiang HS, Chu YL, Chen A, Lin YF, Chung JG. Quercetin induced cell apoptosis and altered gene expression in AGS human gastric cancer cells. ENVIRONMENTAL TOXICOLOGY 2018; 33:1168-1181. [PMID: 30152185 DOI: 10.1002/tox.22623] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 05/20/2023]
Abstract
Quercetin is one of the natural components from natural plant and it induces cell apoptosis in many human cancer cell lines. However, no available reports show that quercetin induces apoptosis and altered associated gene expressions in human gastric cancer cells, thus, we investigated the effect of quercetin on the apoptotic cell death and associated gene expression in human gastric cancer AGS cells. Results indicated that quercetin induced cell morphological changes and reduced total viability via apoptotic cell death in AGS cells. Furthermore, results from flow cytometric assay indicated that quercetin increased reactive oxygen species (ROS) production, decreased the levels of mitochondrial membrane potential (ΔΨm ), and increased the apoptotic cell number in AGS cells. Results from western blotting showed that quercetin decreased anti-apoptotic protein of Mcl-1, Bcl-2, and Bcl-x but increased pro-apoptotic protein of Bad, Bax, and Bid. Furthermore, quercetin increased the gene expressions of TNFRSF10D (Tumor necrosis factor receptor superfamily, member 10d, decoy with truncated death domain), TP53INP1 (tumor protein p53 inducible nuclear protein 1), and JUNB (jun B proto-oncogene) but decreased the gene expression of VEGFB (vascular endothelial growth factor B), CDK10 (cyclin-dependent kinase 10), and KDELC2 (KDEL [Lys-Asp-Glu-Leu] containing 2) that are associated with apoptosis pathways. Thus, those findings may offer more information regarding the molecular, gene expression, and signaling pathway for quercetin induced apoptotic cell death in human gastric cancer cells.
Collapse
Affiliation(s)
- Hung-Sheng Shang
- Graduate Institute of Clinical of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsu-Feng Lu
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan
- Department of Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Ching-Hsiao Lee
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Han-Sun Chiang
- Graduate Institute of Basic Medicine, Fu-Jen Catholic University, New Taipei city, Taiwan
| | - Yung-Lin Chu
- International Master's Degree Program in Food Science, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Ann Chen
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuh-Feng Lin
- Graduate Institute of Clinical of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Shuang Ho Hospital, New Taipei City, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
16
|
Down-regulation of Skp2 expression inhibits invasion and lung metastasis in osteosarcoma. Sci Rep 2018; 8:14294. [PMID: 30250282 PMCID: PMC6155331 DOI: 10.1038/s41598-018-32428-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS), the most common primary cancer of bone, exhibits a high propensity for local invasion and distant metastasis. This study sought to elucidate the role of S phase kinase-associated protein (Skp2) in osteosarcoma invasion and metastasis and to explore flavokawain A (FKA), a natural chalcone from kava extract, as a potential Skp2 targeting agent for preventing osteosarcoma progression. Skp2 was found to be overexpressed in multiple osteosarcoma cell lines, including 5 standard and 8 primary patient-derived cell lines. Patients whose tumors expressed high levels of Skp2 sustained a significantly worse metastasis-free (p = 0.0095) and overall survival (p = 0.0013) than those with low Skp2. Skp2 knockdown markedly reduced in vitro cellular invasion and in vivo lung metastasis in an orthotopic mouse model of osteosarcoma. Similar to Skp2 knockdown, treatment with FKA also reduced Skp2 expression in osteosarcoma cell lines and blocked the invasion of osteosarcoma cells in vitro and lung metastasis in vivo. Together, our findings suggest that Skp2 is a promising therapeutic target in osteosarcoma, and that FKA may be an effective Skp2-targeted therapy to reduce osteosarcoma metastasis.
Collapse
|
17
|
Liu L, Liu K, Yan Y, Chu Z, Tang Y, Tang C. Two Transcripts of FBXO5 Promote Migration and Osteogenic Differentiation of Human Periodontal Ligament Mesenchymal Stem Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7849294. [PMID: 29850565 PMCID: PMC5933072 DOI: 10.1155/2018/7849294] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/03/2018] [Accepted: 03/13/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Enhanced migration and osteogenic differentiation of mesenchymal stem cells (MSCs) are beneficial for MSC-mediated periodontal tissue regeneration, a promising method for periodontitis treatment. FBXO5, a member of the F-box protein family, is involved in the osteogenic differentiation of MSCs. Here, we investigated the effect of FBXO5 on human periodontal ligament stem cells (hPDLSCs). MATERIALS AND METHODS hPDLSCs were isolated from periodontal ligament tissue. Lentivirus FBXO5 shRNA was used to silence FBXO5 expression. Two transcripts of FBXO5 were overexpressed and transduced into hPDLSCs via retroviral infection. Migration and osteogenic differentiation of hPDLSCs were evaluated using the scratch migration assay, alkaline phosphatase (ALP) activity, ALP staining, alizarin red staining, western blotting, and real-time polymerase chain reaction. RESULTS The expression of FBXO5 was upregulated after osteogenic induction in hPDLSCs. FBXO5 knockdown attenuated migration, inhibited ALP activity and mineralization, and decreased RUNX2, OSX, and OCN expression, while the overexpression of two transcript isoforms significantly accelerated migration, enhanced ALP activity and mineralization, and increased RUNX2, OSX, and OCN expression in hPDLSCs. CONCLUSIONS Both isoforms of FBXO5 promoted the migration and osteogenic differentiation potential of hPDLSCs, which identified a potential target for improving periodontal tissue regeneration.
Collapse
Affiliation(s)
- Lin Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kun Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanzhe Yan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhuangzhuang Chu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Tang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunbo Tang
- Department of Dental Implantology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Ding L, Wang C, Cui Y, Han X, Zhou Y, Bai J, Li R. S-phase kinase-associated protein 2 is involved in epithelial-mesenchymal transition in methotrexate-resistant osteosarcoma cells. Int J Oncol 2018; 52:1841-1852. [PMID: 29620168 PMCID: PMC5919717 DOI: 10.3892/ijo.2018.4345] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/23/2018] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS), a common worldwide primary aggressive bone malignancy, arises from primitive transformed cells of mesenchymal origin and usually attacks adolescents and young adults. Methotrexate (MTX) is the anti-folate drug used as a pivotal chemotherapeutic agent in the treatment of OS. However, patients with OS often develop drug resistance, leading to poor treatment outcomes. In the present study, in order to explore the underlying mechanisms responsible for MTX resistance, we established MTX-resistant OS cells using the U2OS and MG63 cell lines and examined whether MTX-resistant OS cells underwent epithelial-mesenchymal transition (EMT) by Transwell assay, wound healing assay, MTT assay, RT-PCR and western blot analysis. We found that the viability of the MTX-resistant cells remained relatively unaltered following further treatment with MTX compared to the parental cells. The resistant cells appeared to possess a mesenchymal phenotype, with an elongated and more spindle-like shape, and acquired enhanced invasive, migratory and attachment abilities. The measurement of EMT markers also supported EMT transition in the MTX-resistant OS cells. Our result further demonstrated that the overexpression of S-phase kinase-associated protein 2 (Skp2) was closely involved in the resistance of OS cells to MTX and in the acquirement of EMT properties. Thus, the pharmacological inhibition of Skp2 may prove to be a novel therapeutic strategy with which to overcome drug resistance in OS.
Collapse
Affiliation(s)
- Lu Ding
- Department of Orthopedics, Fifth Affiliated Hospital, Xinjiang Medical , Urumqi, Xinjiang 830011, P.R. China
| | - Chengwei Wang
- Department of Orthopedics, Sixth Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830002, P.R. China
| | - Yong Cui
- Department of Orthopedics, Fifth Affiliated Hospital, Xinjiang Medical , Urumqi, Xinjiang 830011, P.R. China
| | - Xiaoping Han
- Department of Orthopedics, Fifth Affiliated Hospital, Xinjiang Medical , Urumqi, Xinjiang 830011, P.R. China
| | - Yang Zhou
- Department of Orthopedics, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Jingping Bai
- Department of Orthopedics, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Rong Li
- Department of Maternal, Child and Adolescent Health, College of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
19
|
Abstract
Pim kinases are being implicated in oncogenic process in various human cancers. Pim kinases primarily deal with three broad categories of functions such as tumorigenesis, protecting cells from apoptotic signals and evading immune attacks. Here in this review, we discuss the regulation of Pim kinases and their expression, and how these kinases defend cancer cells from therapeutic and immune attacks with special emphasis on how Pim kinases maintain their own expression during apoptosis and cellular transformation, defend mitochondria during apoptosis, defend cancer cells from immune attack, defend cancer cells from therapeutic attack, choose localization, self-regulation, activation of oncogenic transcription, metabolic regulation and so on. In addition, we also discuss how Pim kinases contribute to tumorigenesis by regulating cellular transformation and glycolysis to reinforce the importance of Pim kinases in cancer and cancer stem cells.
Collapse
|
20
|
Bu W, Luo T. miR-1297 Promotes Cell Proliferation of Non-Small Cell Lung Cancer Cells: Involving in PTEN/Akt/Skp2 Signaling Pathway. DNA Cell Biol 2017; 36:976-982. [PMID: 28872922 DOI: 10.1089/dna.2017.3886] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Wenjin Bu
- Chongqing Medical University, Chongqing, China
- Department of Ultrasonic Medicine, Inner Mongolia People's Hospital, Hohhot, China
| | - Tianyou Luo
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Duan X, Bai J, Wei J, Li Z, Liu X, Xu G. MicroRNA-508-5p suppresses metastasis in human gastric cancer by targeting S-phase kinase‑associated protein 2. Mol Med Rep 2017. [PMID: 28627698 DOI: 10.3892/mmr.2017.6793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
S-phase kinase-associated protein 2 (SKP2), a potent oncogene was revealed to be upregulated in gastric cancer (GC) tissue samples, in which SKP2 was inversely correlated with microRNA (miR)‑508‑5p transcripts. In present study, the functional effect of miR‑508‑5p on SKP2 and its metastatic potential were investigated in SGC‑7901 GC cells. Significant downregulation of the miR‑508‑5p transcript was associated with the progression of GC. Furthermore, the overexpression of miR‑508‑5p was demonstrated to inhibit the proliferation, migration and invasion of SGC‑7901 cells, as well as induced cell apoptosis and cell cycle arrest at the G0/G1 phase in vitro. The overexpression of miR‑508‑5p was able to downregulate the expression of the SKP2 oncogene, through a mechanism by which miR‑508‑5p directly targeted the SKP2 gene. Thus, regulating transcriptional and post‑transcriptional SKP2 expression, as demonstrated using luciferase reporter assays, reverse transcription‑quantitative polymerase chain reaction analysis and immunoblotting assays. The results of the present study identified that miR‑508‑5p functionally affects the SKP2 gene and reduces metastatic potential in GC, suggesting a novel role of miR‑508‑5p in the regulation of SKP2 and cell cycle.
Collapse
Affiliation(s)
- Xiangguo Duan
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jing Bai
- Department of Laboratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jun Wei
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Zhenhao Li
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xinlan Liu
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Guangxian Xu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
22
|
Overexpression of SKP2 Inhibits the Radiation-Induced Bystander Effects of Esophageal Carcinoma. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14020155. [PMID: 28178195 PMCID: PMC5334709 DOI: 10.3390/ijerph14020155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/19/2017] [Accepted: 01/31/2017] [Indexed: 01/29/2023]
Abstract
Background: To investigate the effects of S-phase kinase protein 2 (SKP2) expression on the radiation induced bystander effect (RIBE) in esophageal cancer (EC) cells. Materials and Methods: Western blot was used to detect the levels of SKP2, Rad51, and Ku70 in EC cells. Positive transfection, RNAi, micronucleus (MN), and γ-H2AX focus formation assay were used to investigate the effects of SKP2 on RIBE induced by irradiated cells. Results: We found a significant negative correlation between SKP2 expression and MN frequency (p < 0.05) induced by RIBE. The results were further confirmed by positive transfection, RNAi, and rescue experiments.γ-H2AX focus formation assay results indicated that overexpression of SKP2 in the irradiated cells inhibited the DNA damage of RIBE cells. However, when SKP2 expression decreased in irradiated cells, the DNA damage of RIBE cells increased. Increased or decreased expression levels of SKP2 had effects on Rad51 expression under the conditions of RIBE. Conclusions: These results showed, for the first time, that SKP2 expression can inhibit RIBE of EC cells. The mechanism may function, at least partly, through the regulation of Rad51 in the ability to repair DNA damage.
Collapse
|
23
|
Bonnet A, Servin B, Mulsant P, Mandon-Pepin B. Spatio-Temporal Gene Expression Profiling during In Vivo Early Ovarian Folliculogenesis: Integrated Transcriptomic Study and Molecular Signature of Early Follicular Growth. PLoS One 2015; 10:e0141482. [PMID: 26540452 PMCID: PMC4634757 DOI: 10.1371/journal.pone.0141482] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/08/2015] [Indexed: 11/19/2022] Open
Abstract
Background The successful achievement of early ovarian folliculogenesis is important for fertility and reproductive life span. This complex biological process requires the appropriate expression of numerous genes at each developmental stage, in each follicular compartment. Relatively little is known at present about the molecular mechanisms that drive this process, and most gene expression studies have been performed in rodents and without considering the different follicular compartments. Results We used RNA-seq technology to explore the sheep transcriptome during early ovarian follicular development in the two main compartments: oocytes and granulosa cells. We documented the differential expression of 3,015 genes during this phase and described the gene expression dynamic specific to these compartments. We showed that important steps occurred during primary/secondary transition in sheep. We also described the in vivo molecular course of a number of pathways. In oocytes, these pathways documented the chronology of the acquisition of meiotic competence, migration and cellular organization, while in granulosa cells they concerned adhesion, the formation of cytoplasmic projections and steroid synthesis. This study proposes the involvement in this process of several members of the integrin and BMP families. The expression of genes such as Kruppel-like factor 9 (KLF9) and BMP binding endothelial regulator (BMPER) was highlighted for the first time during early follicular development, and their proteins were also predicted to be involved in gene regulation. Finally, we selected a data set of 24 biomarkers that enabled the discrimination of early follicular stages and thus offer a molecular signature of early follicular growth. This set of biomarkers includes known genes such as SPO11 meiotic protein covalently bound to DSB (SPO11), bone morphogenetic protein 15 (BMP15) and WEE1 homolog 2 (S. pombe)(WEE2) which play critical roles in follicular development but other biomarkers are also likely to play significant roles in this process. Conclusions To our knowledge, this is the first in vivo spatio-temporal exploration of transcriptomes derived from early follicles in sheep.
Collapse
Affiliation(s)
- Agnes Bonnet
- INRA, UMR 1388 GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31076 Toulouse, France
- * E-mail:
| | - Bertrand Servin
- INRA, UMR 1388 GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31076 Toulouse, France
| | - Philippe Mulsant
- INRA, UMR 1388 GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31076 Toulouse, France
| | - Beatrice Mandon-Pepin
- INRA, UMR1198 Biologie du Développement et de la Reproduction, F-78350 Jouy-en-Josas, France
| |
Collapse
|
24
|
Chen P, Yao GD. The role of cullin proteins in gastric cancer. Tumour Biol 2015; 37:29-37. [PMID: 26472722 DOI: 10.1007/s13277-015-4154-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/23/2015] [Indexed: 01/09/2023] Open
Abstract
The cullin proteins are a family of scaffolding proteins that associate with RING proteins and ubiquitin E3 ligases and mediate substrate-receptor bindings. Thus, cullin proteins regulate the specificity of ubiquitin targeting in the regulation of proteins involved in various cellular processes, including proliferation, differentiation, and apoptosis. There are seven cullin proteins that have been identified in eukaryotes: CUL1, CUL2, CUL3, CUL4A, CUL4B, CUL5, and CUL7/p53-associated parkin-like cytoplasmic protein. All of these proteins contain a conserved cullin homology domain that binds to RING box proteins. Cullin-RING ubiquitin ligase complexes are activated upon post-translational modification by neural precursor cell-expressed, developmentally downregulated protein 8. The aberrant expression of several cullin proteins has been implicated in many cancers though the significance in gastric cancer has been less well investigated. This review provides the first systematic discussion of the associations between all members of the cullin protein family and gastric cancer. Functional and regulatory mechanisms of cullin proteins in gastric carcinoma progression are also summarized along with a discussion concerning future research areas. Accumulating evidence suggests a critical role of cullin proteins in tumorigenesis, and a better understanding of the function of these individual cullin proteins and their targets will help identify potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Peng Chen
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Tong-Dao-Bei Street, Hohhot, Inner Mongolia, 010050, People's Republic of China
| | - Guo-Dong Yao
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Tong-Dao-Bei Street, Hohhot, Inner Mongolia, 010050, People's Republic of China.
| |
Collapse
|
25
|
Zhai B, Hu F, Yan H, Zhao D, Jin X, Fang T, Pan S, Sun X, Xu L. Bufalin Reverses Resistance to Sorafenib by Inhibiting Akt Activation in Hepatocellular Carcinoma: The Role of Endoplasmic Reticulum Stress. PLoS One 2015; 10:e0138485. [PMID: 26381511 PMCID: PMC4575108 DOI: 10.1371/journal.pone.0138485] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 08/31/2015] [Indexed: 12/17/2022] Open
Abstract
Sorafenib is the standard first-line therapeutic treatment for patients with advanced hepatocellular carcinoma (HCC), but its use is hampered by the development of drug resistance. The activation of Akt by sorafenib is thought to be responsible for this resistance. Bufalin is the major active ingredient of the traditional Chinese medicine Chan su, which inhibits Akt activation; therefore, Chan su is currently used in the clinic to treat cancer. The present study aimed to investigate the ability of bufalin to reverse both inherent and acquired resistance to sorafenib. Bufalin synergized with sorafenib to inhibit tumor cell proliferation and induce apoptosis. This effect was at least partially due to the ability of bufalin to inhibit Akt activation by sorafenib. Moreover, the ability of bufalin to inactivate Akt depended on endoplasmic reticulum (ER) stress mediated by inositol-requiring enzyme 1 (IRE1). Silencing IRE1 with siRNA blocked the bufalin-induced Akt inactivation, but silencing eukaryotic initiation factor 2 (eIF2) or C/EBP-homologous protein (CHOP) did not have the same effect. Additionally, silencing Akt did not influence IRE1, CHOP or phosphorylated eIF2α expression. Two sorafenib-resistant HCC cell lines, which were established from human HCC HepG2 and Huh7 cells, were refractory to sorafenib-induced growth inhibition but were sensitive to bufalin. Thus, Bufalin reversed acquired resistance to sorafenib by downregulating phosphorylated Akt in an ER-stress-dependent manner via the IRE1 pathway. These findings warrant further studies to examine the utility of bufalin alone or in combination with sorafenib as a first- or second-line treatment after sorafenib failure for advanced HCC.
Collapse
Affiliation(s)
- Bo Zhai
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fengli Hu
- Department of Gastroenterology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Haijiang Yan
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dali Zhao
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xin Jin
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Taishi Fang
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shangha Pan
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xueying Sun
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lishan Xu
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- * E-mail:
| |
Collapse
|
26
|
The novel protective role of P27 in MLN4924-treated gastric cancer cells. Cell Death Dis 2015; 6:e1867. [PMID: 26313918 PMCID: PMC4558507 DOI: 10.1038/cddis.2015.215] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/12/2015] [Accepted: 06/16/2015] [Indexed: 12/19/2022]
Abstract
The tumor-suppressor gene cyclin-dependent kinase inhibitor 1B (P27) is downregulated in gastric cancer cells mainly through proteolytic degradation mediated by the SKP-Cullin1-F-Box (SCF) complex. But the correlation between its downregulation and gastric cancer prognosis still remains indefinite. MLN4924, an anti-tumor agent, which suppresses the SCF complex by inhibiting Cullin1 neddylation, emerges as a promising tool to elucidate its functions in gastric cancer cells. In this study, MLN4924 induced significant growth inhibition of gastric cancer cells in a dose-dependent manner, along with the simultaneous accumulation of P27 and cell cycle abnormalities such as G2/M arrest. Importantly, we found that P27 silencing in MLN4924-treated cells resulted in an enhancement of growth inhibition both in vitro and in vivo. Mechanism analysis revealed the antagonism effects of antioxidants to this excess apoptosis, suggesting reactive oxygen species (ROS) overproduction especially in the mitochondria was the principal cause of the augmentation. Moreover, the robust ROS attacked the mitochondria to initiate collapse of the mitochondrial membrane permeability and the exportation of apoptosis-inducing factor (AIF), IAP-binding mitochondrial protein (SMAC/DIABLO) and cytochrome c. Finally, we also found that P27 knockdown affected the expression profile of several critical BH3 family members to amplify the mitochondrial dysfunction and apoptosis. In summary, our findings unveiled a protective role of P27 by maintaining mitochondrial membrane permeability in MLN4924-treated gastric cancer cells, and therefore highlighted the potential combination of MLN4924 with P27 inhibition to improve its therapeutic efficacy.
Collapse
|
27
|
Wang J, Huang Y, Guan Z, Zhang JL, Su HK, Zhang W, Yue CF, Yan M, Guan S, Liu QQ. E3-ligase Skp2 predicts poor prognosis and maintains cancer stem cell pool in nasopharyngeal carcinoma. Oncotarget 2015; 5:5591-601. [PMID: 25015320 PMCID: PMC4170633 DOI: 10.18632/oncotarget.2149] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the severe head and neck carcinomas, which is rare in west countries but has high incidence in Southern Asia especially South China. Although NPC is relatively sensitive to radiotherapy, the prognosis of patients is poor due to the advanced stage at the time of diagnosis. Therefore, it is important to understand the mechanisms involved in tumorigenesis and develop early diagnostic techniques. S-phase kinase associated protein 2 (Skp2) is overexpressed in several human cancers and associates with poor prognosis. However, its function in NPC has not been fully addressed. In this study we found Skp2 was highly expressed in NPC specimen and correlated with poor prognosis. We generated Skp2 knockdown cells to further delineate its role in NPC development. Knockdown of Skp2 partially reduced cell proliferation, promoted cellular senescence, and decreased the population of stem cell like aldehyde dehydrogenase1 positive cells as well as their self-renewal ability. Our study not only interprets the predictive role of Skp2 in the poor prognosis of NPC patients, but also reveals that Skp2 regulates the NPC cancer stem cell maintenance, which shed lights on the target therapy and early diagnosis of NPC in clinical application.
Collapse
Affiliation(s)
- Jing Wang
- State key laboratory of oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China. Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Huang
- State key laboratory of oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhong Guan
- Department of Otorhinolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jia-Liang Zhang
- Department of Otorhinolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong-Kai Su
- State key laboratory of oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Zhang
- State key laboratory of oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China. Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Cai-Feng Yue
- State key laboratory of oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China. Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Min Yan
- State key laboratory of oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China. Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Su Guan
- School of Bioscience and Biotechnology, South China University of Technology, Guangzhou, China
| | - Quentin Qiang Liu
- State key laboratory of oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China. Department of Research Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
28
|
LIN LELE, JIANG HONGPENG, HUANG MINGKUI, HOU XU, SUN XUEPU, JIANG XIAN, DONG XUESONG, SUN XUEYING, ZHOU BAOGUO, QIAO HAIQUAN. Depletion of histone deacetylase 1 inhibits metastatic abilities of gastric cancer cells by regulating the miR-34a/CD44 pathway. Oncol Rep 2015; 34:663-72. [DOI: 10.3892/or.2015.4010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/02/2015] [Indexed: 11/05/2022] Open
|
29
|
GONG JIAN, CAO JUAN, LIU GUINAN, HUO JIRONG. Function and mechanism of F-box proteins in gastric cancer (Review). Int J Oncol 2015; 47:43-50. [DOI: 10.3892/ijo.2015.2983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/24/2015] [Indexed: 11/06/2022] Open
|
30
|
Wei WY, Yan LH, Wang XT, Li L, Cao WL, Zhang XS, Zhan ZX, Yu H, Xie YB, Xiao Q. E2F-1 overexpression inhibits human gastric cancer MGC-803 cell growth in vivo. World J Gastroenterol 2015; 21:491-501. [PMID: 25593464 PMCID: PMC4292280 DOI: 10.3748/wjg.v21.i2.491] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/16/2014] [Accepted: 07/16/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the influence of E2F-1 on the growth of human gastric cancer (GC) cells in vivo and the mechanism involved.
METHODS: E2F-1 recombinant lentiviral vectors were injected into xenograft tumors of MGC-803 cells in nude mice, and then tumor growth was investigated. Overexpression of transcription factor E2F-1 was assessed by reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting analysis. Apoptosis rates were determined using a terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay. Expression levels of certain cell cycle regulators and apoptosis-related proteins, such as Bax, survivin, Bcl-2, cyclin D1, S-phase kinase-associated protein 2, and c-Myc were examined by Western blotting and RT-PCR.
RESULTS: Xenograft tumors of MGC-803 cells in nude mice injected with E2F-1 recombinant lentiviral vectors stably overexpressed the E2F-1 gene as measured by semi-quantitative RT-PCR (relative mRNA expression: 0.10 ± 0.02 vs 0.05 ± 0.02 for control vector and 0.06 ± 0.03 for no infection; both P < 0.01) and Western blotting (relative protein expression: 1.90 ± 0.05 vs 1.10 ± 0.03 in control vector infected and 1.11 ± 0.02 for no infection; both P < 0.01). The growth-curve of tumor volumes revealed that infection with E2F-1 recombinant lentiviral vectors significantly inhibited the growth of human GC xenografts (2.81 ± 1.02 vs 6.18 ± 1.15 in control vector infected and 5.87 ± 1.23 with no infection; both P < 0.05) at 15 d after treatment. TUNEL analysis demonstrated that E2F-1 overexpression promoted tumor cell apoptosis (18.6% ± 2.3% vs 6.7% ± 1.2% in control vector infected 6.3% ± 1.2% for no infection; both P < 0.05). Furthermore, lentiviral vector-mediated E2F-1 overexpression increased the expression of Bax and suppressed survivin, Bcl-2, cyclin D1, Skp2, and c-Myc expression in tumor tissue.
CONCLUSION: E2F-1 inhibits growth of GC cells via regulating multiple signaling pathways, and may play an important role in targeted therapy for GC.
Collapse
|
31
|
Qi M, Liu D, Zhang S, Hu P, Sang T. Inhibition of S-phase kinase-associated protein 2-mediated p27 degradation suppresses tumorigenesis and the progression of hepatocellular carcinoma. Mol Med Rep 2015; 11:3934-40. [PMID: 25572801 DOI: 10.3892/mmr.2015.3156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 10/01/2014] [Indexed: 11/06/2022] Open
Abstract
In order to determine the protein expression of S‑phase kinase‑associated protein 2 (Skp2) and p27kip1, and to evaluate their possible prognostic values in malignant liver cancer, tissue samples from 50 patients and 40 controls were assessed and analyzed by immunohistochemistry and western blot analysis. Positive expression of Skp2 was observed in 35 (70.0%) of the hepatocellular carcinoma samples; however, the positive expression of p27kip1 was observed in 6 (15.0%) of the hepatocellular carcinoma samples. The expression of Skp2 was significantly negatively correlated with the expression of p27 (P<0.01). The results from Annexin V‑propidium iodide staining and MTT assays indicated that interference of Skp2 significantly induced apoptosis and inhibited the proliferation of SSMC‑7721 cells. In addition, the levels of endogenous p27 increased in the HepG2 and SSMC‑7721 cells following transfection with siRNA specific to Skp2, suggesting that the Skp2‑mediated degradation of p27kip1 was important in the proliferation of tumor cells. The present study, therefore, provided a molecular reference for the treatment of liver cancer.
Collapse
Affiliation(s)
- Ming Qi
- Department of Digestive System, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Dongmei Liu
- Department of Transfusion Center, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Shuhong Zhang
- Department of Digestive System, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Peixin Hu
- Department of Digestive System, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Tan Sang
- Department of Hematology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
32
|
Zhai B, Jiang X, He C, Zhao D, Ma L, Xu L, Jiang H, Sun X. Arsenic trioxide potentiates the anti-cancer activities of sorafenib against hepatocellular carcinoma by inhibiting Akt activation. Tumour Biol 2014; 36:2323-34. [PMID: 25416439 DOI: 10.1007/s13277-014-2839-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/11/2014] [Indexed: 01/05/2023] Open
Abstract
Sorafenib is the standard first-line systemic drug for advanced hepatocellular carcinoma (HCC), but it also induces the activation of Akt, which contributes to the mechanisms for the resistance to sorafenib. Arsenic trioxide (ATO) is a currently clinically used anticancer drug and displays its anticancer activities by inhibiting Akt activation. Therefore, we hypothesized that ATO may potentiate the anti-cancer activities of sorafenib against HCC. The results have demonstrated that ATO synergized with sorafenib to inhibit the proliferation and promote the apoptosis of HCC cells by diminishing the increased activation of Akt by sorafenib. ATO was shown to inhibit the expression or activation of Akt downstream factors, including glycogen synthase kinase (GSK)-3β, mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase (S6K), and eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1), which regulate cell apoptosis and were upregulated or activated by sorafenib. Both sorafenib and ATO downregulated the expression of cyclin D1, resulting in HCC cells arrested at G0/G1 phase. ATO downregulated the expression of Bcl-2 and Bcl-xL and upregulated the expression of Bax, indicating that ATO could induce the apoptosis of HCC cells through the intrinsic pathways; but sorafenib showed little effects on these proteins of Bcl-2 family. ATO synergized with sorafenib to suppress the growth of HCC tumors established in mice by inhibiting the proliferation and inducing the apoptosis of HCC cells in situ. These results indicate that ATO may be a potential agent that given in combination with sorafenib acts synergistically for treating HCC.
Collapse
Affiliation(s)
- Bo Zhai
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Chang R, Wei L, Lu Y, Cui X, Lu C, Liu L, Jiang D, Xiong Y, Wang G, Wan C, Qian H. Upregulated expression of ubiquitin-conjugating enzyme E2Q1 (UBE2Q1) is associated with enhanced cell proliferation and poor prognosis in human hapatocellular carcinoma. J Mol Histol 2014; 46:45-56. [PMID: 25311764 DOI: 10.1007/s10735-014-9596-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/30/2014] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer in the world. Ubiquitin-proteasome system has been shown to play a pivotal role in the pathophysiology of HCC and other malignancies. UBE2Q1 is a putative E2 ubiquitin conjugating enzyme, and may be involved in the regulation of cancer-related proteins. In this study, we investigated the expression pattern of UBE2Q1 in HCC cell lines and human HCC specimens, and its potential clinical and biological significance in HCC. Western blot and immunohistochemical analyses revealed that UBE2Q1 was significantly upregulated in HCC tumorous tissues compared with the adjacent noncancerous ones. Next, univariate and multivariate survival analyses were performed to determine the prognostic significance of UBE2Q1 in HCC. The results showed that upregulated expression of UBE2Q1 was positively correlated with high histological grades of HCC and predicted poor prognosis. In addition, the expression of UBE2Q1 was progressively increased in serum-refed HCC cells. UBE2Q1 depletion by small interfering RNA inhibited cell proliferation and led to G1 phase arrest in HepG2 and BEL-7404 cells. Furthermore, we showed that cells transfected with UBE2Q1-targeting siRNA resulted in significant increase in the levels of p53, p21 in HepG2 and BEL-7404 cells. These data imply that UBE2Q1 is upregulated in liver cancer cell lines and tumorous samples and may play a role in the development of HCC.
Collapse
Affiliation(s)
- Renan Chang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gong J, Lv L, Huo J. Roles of F-box proteins in human digestive system tumors (Review). Int J Oncol 2014; 45:2199-207. [PMID: 25270675 DOI: 10.3892/ijo.2014.2684] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/16/2014] [Indexed: 12/16/2022] Open
Abstract
F-box proteins (FBPs), the substrate-recognition subunit of E3 ubiquitin (Ub) ligase, are the important components of Ub proteasome system (UPS). FBPs are involved in multiple cellular processes through ubiquitylation and subsequent degradation of their target proteins. Many studies have described the roles of FBPs in human cancers. Digestive system tumors account for a large proportion of all the tumors, and their mortality is very high. This review summarizes for the first time the roles of FBPs in digestive system tumorige-nesis and tumor progression, aiming at finding new routes for the rational design of targeted anticancer therapies in digestive system tumors.
Collapse
Affiliation(s)
- Jian Gong
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Liang Lv
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jirong Huo
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
35
|
Lu H, Cao X, Zhang H, Sun G, Fan G, Chen L, Wang S. Imbalance between MMP-2, 9 and TIMP-1 promote the invasion and metastasis of renal cell carcinoma via SKP2 signaling pathways. Tumour Biol 2014; 35:9807-13. [DOI: 10.1007/s13277-014-2256-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/18/2014] [Indexed: 12/21/2022] Open
|
36
|
Chen H, Mo X, Yu J, Huang S, Huang Z, Gao L. Interference of Skp2 effectively inhibits the development and metastasis of colon carcinoma. Mol Med Rep 2014; 10:1129-35. [PMID: 24913024 DOI: 10.3892/mmr.2014.2308] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 04/14/2014] [Indexed: 11/06/2022] Open
Abstract
Colon cancer is a common type of malignancy in the digestive system. The aim of the present study was to investigate the role of S-phase kinase-associated protein 2 (Skp2) in colon carcinoma and to identify whether depletion of Skp2 by Skp2‑RNA interference (RNAi) attenuates the proliferation and migration of colon carcinoma. Three pairs of small interfering (si)RNA were designed based on the Skp2 gene sequence and the most effective one was used to silence the Skp2 gene in SW620 cells. Subsequent to the interference, quantitative polymerase chain reaction and western blot analysis were used for detecting the expression of Skp-2 mRNA and protein, respectively. The data demonstrated that the Skp2‑siRNA effectively inhibited proliferation (P<0.01), increased the levels of apoptosis and induced G0/G1 phase arrest of the SW620 cells (P<0.01). Transfection of the Skp2 siRNA into SW620 cells effectively reduced Skp2 protein levels, while p27 protein levels increased. In the in vivo experiments, a lentiviral vector of the Skp2-RNAi transfected into SW620 cells markedly inhibited Skp2 expression, as detected by immunohistochemical analysis of nude mice. Additionally, tumorigenicity experiments showed that inhibition of Skp2 significantly increased the survival rate of nude mice. Thus, the in vitro and in vivo results demonstrated that interference of Skp2 expression significantly inhibited the proliferation and induced the apoptosis of SW620 cells. This suggests that Skp2 protein has an important role in the progression of colon cancer. Therefore, Skp2 may enable the early diagnosis of colon cancer and provide new insights into molecular targets for cancer therapy.
Collapse
Affiliation(s)
- Haijin Chen
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Xiaodong Mo
- Department of Gastrointestinal Surgery, PLA No. 101 Hospital, Wuxi, Jiangsu 214044, P.R. China
| | - Jinlong Yu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Shuxin Huang
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Liping Gao
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
37
|
Huang MH, Lin SZ, Lin PC, Chiou TW, Harn YW, Ho LI, Chan TM, Chou CW, Chuang CH, Su HL, Harn HJ. Brain tumor senescence might be mediated by downregulation of S-phase kinase-associated protein 2 via butylidenephthalide leading to decreased cell viability. Tumour Biol 2014; 35:4875-84. [PMID: 24464249 DOI: 10.1007/s13277-014-1639-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 01/07/2014] [Indexed: 12/21/2022] Open
Abstract
Developing an effective drug for treating human glioblastoma multiform (GBM) has been investigated persistently. A pure compound butylidenephthalide (BP), isolated from Angelica sinensis, has been shown the activities to arrest the growth and initiate apoptosis of GBM in our previous reports. In this study, we further demonstrated that BP treatment accelerates the cell senescence in a dose-dependent manner in vitro and in vivo. S-phase kinase-associated protein 2 (Skp2), a proto-oncogene, is generally upregulated in cancer. We found that it was downregulated in BP-treated GBM cells. The downregulation of Skp2 is parallel with increasing p16 and p21 expression which causes G0/G1 arrest and tumor cell senescence. We also found that restoring the Skp2 protein level by exogenous overexpression prevents the BP-induced cell senescence. Therefore, the linkage between cell senescence and Skp2 expression is strengthened. Promoter binding analysis further detailed that the BP-mediated SP1 reduction might involve in the Skp2 downregulation. In summary, these results emphasize that BP-triggered senescence in GBM cells is highly associated with its control on Skp2 regulation.
Collapse
Affiliation(s)
- Mao-Hsuan Huang
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Xu SY, Wang F, Wei G, Wang B, Yang JY, Huang YZ, Zhang L, Zheng F, Guo LY, Wang JN, Tang JM. S-phase kinase-associated protein 2 knockdown blocks colorectal cancer growth via regulation of both p27 and p16 expression. Cancer Gene Ther 2013; 20:690-4. [PMID: 24336114 DOI: 10.1038/cgt.2013.70] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 10/17/2013] [Indexed: 11/09/2022]
Abstract
The objective of this study was to determine the role and mechanism of S-phase kinase-associated protein 2 (Skp2) in colorectal cancer cell proliferation and survival both in vitro and in vivo. Adenoviral vector expressing Skp2 short hairpin RNA was transduced into SW480 cells. The effects of Skp2 on cell cycle and survival were assessed by Flow Cytometry. Cell proliferation was analyzed by MTT assay. The expression of cell cycle regulators p16 and p27 were measured by western blot. In vivo, human colorectal cancer was produced by xenograft of cancer cells in nude mouse. Tumor growth inhibitory rate was calculated to generate growth curve. Tumor growth was monitored by examining proliferating cell nuclear antigen expression, whereas tumor cell apoptosis was detected by TdT-mediated dUTP nick-end labeling (TUNEL) staining. Knockdown of Skp2 blocked SW480 tumor cell growth and induced cell apoptosis. Skp2 appeared to be very important for the progression of cell cycle at G1/S phase. In vivo, blockade of Skp2 expression inhibited tumor growth and induced tumor apoptosis. Mechanistically, Skp2 regulated the expression of both p27 and p16 both in vitro and in vivo. The conclusion that we derive from this study is that Skp2 regulates colorectal cancer cell growth by inhibiting the expression of cell cycle regulator p27 and p16.
Collapse
Affiliation(s)
- S-Y Xu
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - F Wang
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - G Wei
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - B Wang
- 1] Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, China [2] Hubei University of Medicine, Shiyan, China
| | - J-Y Yang
- Key Laboratory of Human Embryonic Stem Cell Research of Hubei Province, Hubei University of Medicine, Shiyan, China
| | - Y-Z Huang
- 1] Key Laboratory of Human Embryonic Stem Cell Research of Hubei Province, Hubei University of Medicine, Shiyan, China [2] Department of Cardiology and Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - L Zhang
- Key Laboratory of Human Embryonic Stem Cell Research of Hubei Province, Hubei University of Medicine, Shiyan, China
| | - F Zheng
- Key Laboratory of Human Embryonic Stem Cell Research of Hubei Province, Hubei University of Medicine, Shiyan, China
| | - L-Y Guo
- Key Laboratory of Human Embryonic Stem Cell Research of Hubei Province, Hubei University of Medicine, Shiyan, China
| | - J-N Wang
- Key Laboratory of Human Embryonic Stem Cell Research of Hubei Province, Hubei University of Medicine, Shiyan, China
| | - J-M Tang
- 1] Key Laboratory of Human Embryonic Stem Cell Research of Hubei Province, Hubei University of Medicine, Shiyan, China [2] Department of Physiology, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
39
|
Sun XP, Dong X, Lin L, Jiang X, Wei Z, Zhai B, Sun B, Zhang Q, Wang X, Jiang H, Krissansen GW, Qiao H, Sun X. Up-regulation of survivin by AKT and hypoxia-inducible factor 1α contributes to cisplatin resistance in gastric cancer. FEBS J 2013; 281:115-28. [PMID: 24165223 DOI: 10.1111/febs.12577] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/15/2013] [Accepted: 10/16/2013] [Indexed: 12/23/2022]
Abstract
This study investigated the contribution of survivin and its upstream regulators, AKT and hypoxia-inducible factor 1α (HIF-1α), to the resistance of gastric cancer cells to cisplatin (CDDP). We found that over-expression of survivin increased the resistance of SGC7901 and BGC823 gastric cancer cells to CDDP. Its over-expression abrogated CDDP-induced inhibition of cell proliferation and CDDP-induced cell apoptosis. In contrast, down-regulation of survivin expression using small hairpin RNA (shRNA) vectors and the small-molecule inhibitor YM155, or inhibition of survivin function using a recombinant cell-permeable dominant-negative survivin protein (dNSur9), promoted CDDP-induced apoptosis. CDDP-resistant sub-lines generated from the parental SGC7901 and BGC823 cells by exposure to increasing concentrations of CDDP expressed higher levels of HIF-1α and survivin in response to hypoxia, and higher levels of phosphorylated AKT (pAKT). Specific inhibition of AKT reduced the expression of HIF-1α and survivin, whereas specific inhibition or depletion of HIF-1α reduced survivin expression but had no effect on the expression of phosphorylated AKT. The expression levels of survivin affected the therapeutic efficacy of CDDP in treating gastric tumors in mice. Specific inhibition of survivin, AKT and HIF-1α enhanced the sensitivity of CDDP-resistant cells to CDDP. Specific inhibition of survivin, AKT and HIF-1α synergized with CDDP to suppress the growth of gastric tumors that had been engineered to overexpress survivin. In summary, the results provide evidence that up-regulation of survivin by AKT and HIF-1α contributes to CDDP resistance, indicating that inhibition of these pathways may be a potential strategy for overcoming CDDP resistance in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Xue-Pu Sun
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhang M, Peng L, Qiao ZB, He HT, Zhou Y, Xu Z. Inhibition of the PI3K/Akt signaling pathway inhibits cell proliferation and induces apoptosis in hepatocellular carcinoma cell line HepG2. Shijie Huaren Xiaohua Zazhi 2013; 21:2250-2257. [DOI: 10.11569/wcjd.v21.i23.2250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of the PI3K/Akt signaling pathway inhibitor wortmannin on cell proliferation, apoptosis and expression of pAkt, Skp2 and P27kip1 in human hepatocellular carcinoma cell line HepG2.
METHODS: After treatment with different concentrations of wortmannin (0, 10, 50, 100, 200 nmol/L) for different durations (3, 10, 24 h), proliferation of HepG2 cells was analyzed by MTT assay, cell cycle and apoptosis were detected by flow cytometry, expression of pAkt, Skp2 and P27kip1 proteins was detected by Western blot, and the mRNA expression of Skp2 and P27kip1 was detected by reverse transcription-polymerase chain reaction.
RESULTS: Wortmannin inhibited the proliferation of HepG2 cells in a dose- and time-dependent manner. The apoptosis rates of HepG2 cells significantly increased from 8.46% ± 1.17% to 28.03% ± 2.67% after treatment with wortmannin (P < 0.05). Wortmannin induced an increase in the percentage of cells in G0/G1 phase and a decrease in the percentage of cells in S phase cells (both P < 0.05). After treatment with wortmannin, the expression of pAkt and Skp2 proteins was down-regulated, while that of P27kip1 protein was up-regulated (all P < 0.05). In addition, Skp2 mRNA expression in HepG2 cells was significantly down-regulated (P < 0.05), although the expression of P27kip1 mRNA was not changed.
CONCLUSION: Wortmannin inhibits cell proliferation and induces apoptosis in human hepatocellular carcinoma cell line HepG2 possibly by regulating the expression of Skp2 and P27kip1.
Collapse
|
41
|
Ju Y, Yu A, Sun X, Wu D, Zhang H. Glucosamine, a naturally occurring amino monosaccharide, inhibits A549 and H446 cell proliferation by blocking G1/S transition. Mol Med Rep 2013; 8:794-8. [PMID: 23846431 DOI: 10.3892/mmr.2013.1584] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 07/02/2013] [Indexed: 01/14/2023] Open
Abstract
Uncontrolled proliferation is important in tumorigenesis. In the present study, the effects of glucosamine on lung cancer cell proliferation were investigated. The expression of cyclin E, one of the key cyclins in the G1/S transition, and Skp2, the ubiquitin ligase subunit that targets the negative cell cycle regulator, p27Kip1, were also assessed. Moreover, the underlying mechanisms of action of glucosamine were investigated in lung cancer cells. A549 and H446 cells were synchronized using thymidine in the presence or absence of glucosamine. The effect of glucosamine on lung cancer cell proliferation was determined by MTT assay. Cyclin E and p27Kip1 proteins and their phosphorylation levels were detected by western blot analysis. Furthermore, the effect of glucosamine on the cell cycle was evaluated by flow cytometry. Glucosamine was found to inhibit lung cancer cell proliferation and to suppress Skp2 and cyclin E expression. Notably, the phosphorylation levels of cyclin E (Thr62) and p27Kip1 (Thr187) were downregulated by glucosamine, and negatively correlated with degradation. Glucosamine was also found to arrest lung cancer cells in the G1/S phase. Thus, glucosamine may inhibit lung cancer cell proliferation by blocking G1/S transition through the inhibition of cyclin E and Skp2 protein expression.
Collapse
Affiliation(s)
- Yinghua Ju
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | | | | | | | | |
Collapse
|