1
|
Cheng J, Xu L, Xuan Y, Zhou F, Huang A, Zeng S, Wang H, Wang Y, Zhan Y, Yan X, Luo S, Liu Y, Cheng M. Linear polyubiquitylation of Gli protein regulates its protein stability and facilitates tumor growth in colorectal cancer. Cell Death Discov 2024; 10:369. [PMID: 39164252 PMCID: PMC11335874 DOI: 10.1038/s41420-024-02147-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
The linear ubiquitin chain assembly complex (LUBAC) mediates the linear ubiquitination of various proteins and is involved in NF-κB signaling and immune regulation. However, the function and mechanism of linear ubiquitination in regulating oncogenic signaling and tumor growth have remained poorly understood. Herein, we identified Gli proteins, key transcription factors in the Hedgehog (Hh) signaling pathway, as novel substrates of LUBAC. Linear ubiquitination stabilizes Gli proteins, leading to the noncanonical activation of Hh signaling in CRC cells. Furthermore, LUBAC facilitates tumor growth in CRC cells. Additionally, elevated expression of LUBAC components in CRC tissues was observed, and higher expression levels of these components correlated with poor prognosis in CRC patients. Interestingly, inhibition of LUBAC using either a small molecule agonist or RNA silencing specifically suppressed cell growth in CRC cells but had no effect on normal intestinal cells. Taken together, aberrant expression of LUBAC components activates Hh signaling noncanonically by mediating linear ubiquitination, promoting tumor growth in CRC, demonstrating the novel function of linear ubiquitination in regulating the protein stability of its substrates and highlighting the potential of targeting LUBAC as a therapeutic strategy in CRC.
Collapse
Affiliation(s)
- Junyao Cheng
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Linlin Xu
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Pathology and Institute of Molecular Pathology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yanlu Xuan
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Feifei Zhou
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Aidi Huang
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Pathology and Institute of Molecular Pathology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shaopeng Zeng
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hailong Wang
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Centre, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yiting Wang
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuan Zhan
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Pathology and Institute of Molecular Pathology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaohua Yan
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shiwen Luo
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, Department of Pathology and Institute of Molecular Pathology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Minzhang Cheng
- Center for Experimental Medicine, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
2
|
Qiu Z, He S, Lu B, Sun Y, Zhang T, Lv W, Shen D. The E3 ubiquitin ligase RNF135 modulates chemotherapy resistance to oxaliplatin for colorectal cancer by modulating autophagy. Tissue Cell 2024; 86:102282. [PMID: 38056362 DOI: 10.1016/j.tice.2023.102282] [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: 08/04/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND RING finger protein 135 plays an important role in tumorigenesis and is associated with drug resistance. METHODS Bioinformatics analysis showed that RNF135 was significantly differentially expressed in colorectal cancer. RT-qPCR and western blot were used to detect the expression of RNF135. Immunohistochemical analysis were used to measure the expression of RNF135 and Ki-67. RESULTS The expression of RNF135 was up-regulated in human tissue samples and colorectal cancer and was positively correlated with Ki-67. Compared with oxaliplatin sensitive patients, RNF135 expression levels were higher in the tissue of resistant patients. The regulatory effect of RNF135 on colorectal cancer cells was further investigated in vitro. Therefore, inhibition of autophagy by down-regulating RNF135 can partially increase its susceptibility to oxaliplatin.
Collapse
Affiliation(s)
- Zhen Qiu
- Department of Pathology, Hongze People's Hospital, 102 Dongfeng Road, Hongze 223100, China
| | - Shuyan He
- Department of Tumor Center, Affiliated Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin 214400, China
| | - Boyi Lu
- Department of Tumor Center, Affiliated Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin 214400, China
| | - Yuejun Sun
- Department of Pathology, Affiliated Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin 214400, China
| | - Ting Zhang
- Department of Central Laboratory, Affiliated Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin 214400, China
| | - Wei Lv
- Department of Pharmacy, Jiangyin Hospital Affiliated to Nantong University, 163 Shoushan Road, Jiangyin 214400, China.
| | - Dong Shen
- Department of Tumor Center, Affiliated Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin 214400, China.
| |
Collapse
|
3
|
Hoshino K, Nakazawa S, Yokobori T, Hagiwara K, Ishii N, Tsukagoshi M, Igarashi T, Araki K, Harimoto N, Tokunaga F, Shirabe K. RNF31 promotes proliferation and invasion of hepatocellular carcinoma via nuclear factor kappaB activation. Sci Rep 2024; 14:346. [PMID: 38172174 PMCID: PMC10764851 DOI: 10.1038/s41598-023-50594-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
RNF31 is a multifunctional RING finger protein implicated in various inflammatory diseases and cancers. It functions as a core component of the linear ubiquitin chain assembly complex (LUBAC), which activates the nuclear factor kappaB (NF-κB) pathway via the generation of the Met1-linked linear ubiquitin chain. We aimed to clarify the role of RNF31 in the pathogenesis of hepatocellular carcinoma (HCC) and its relevance as a therapeutic target. High RNF31 expression in HCC, assessed by both immunohistochemistry and mRNA levels, was related to worse survival rates among patients with HCC. In vitro experiments showed that RNF31 knockdown in HCC cell lines led to decreased cell proliferation and invasion, as well as suppression of tumor necrosis factor (TNF)-α-induced NF-κB activation. Treatment with HOIPIN-8, a specific LUBAC inhibitor that suppresses RNF31 ubiquitin ligase (E3) activity, showed similar effects, resulting in decreased cell proliferation and invasion. Our clinical and in vitro data showed that RNF31 is a prognostic factor for HCC that promotes tumor aggressiveness via NF-κB activation.
Collapse
Affiliation(s)
- Kouki Hoshino
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Seshiru Nakazawa
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan.
| | | | - Kei Hagiwara
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Norihiro Ishii
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Mariko Tsukagoshi
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Takamichi Igarashi
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Kenichiro Araki
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Norifumi Harimoto
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Fuminori Tokunaga
- Department of Medical Biochemistry, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| |
Collapse
|
4
|
Li J, Liu S, Li S. Mechanisms underlying linear ubiquitination and implications in tumorigenesis and drug discovery. Cell Commun Signal 2023; 21:340. [PMID: 38017534 PMCID: PMC10685518 DOI: 10.1186/s12964-023-01239-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/19/2023] [Indexed: 11/30/2023] Open
Abstract
Linear ubiquitination is a distinct type of ubiquitination that involves attaching a head-to-tail polyubiquitin chain to a substrate protein. Early studies found that linear ubiquitin chains are essential for the TNFα- and IL-1-mediated NF-κB signaling pathways. However, recent studies have discovered at least sixteen linear ubiquitination substrates, which exhibit a broader activity than expected and mediate many other signaling pathways beyond NF-κB signaling. Dysregulation of linear ubiquitination in these pathways has been linked to many types of cancers, such as lymphoma, liver cancer, and breast cancer. Since the discovery of linear ubiquitin, extensive effort has been made to delineate the molecular mechanisms of how dysregulation of linear ubiquitination causes tumorigenesis and cancer development. In this review, we highlight newly discovered linear ubiquitination-mediated signaling pathways, recent advances in the role of linear ubiquitin in different types of cancers, and the development of linear ubiquitin inhibitors. Video Abstract.
Collapse
Affiliation(s)
- Jack Li
- Department of Biosciences, Rice University, Houston, TX, 77005, USA
| | - Sijin Liu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, People's Republic of China.
| | - Shitao Li
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
5
|
Zhang Y, Li QS, Liu HL, Tang HT, Yang HL, Wu DQ, Huang YY, Li LC, Liu LH, Li MX. MKRN1 promotes colorectal cancer metastasis by activating the TGF-β signalling pathway through SNIP1 protein degradation. J Exp Clin Cancer Res 2023; 42:219. [PMID: 37620897 PMCID: PMC10464235 DOI: 10.1186/s13046-023-02788-w] [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: 04/13/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND The Makorin ring finger protein 1 (MKRN1) gene, also called RNF61, is located on the long arm of chromosome 7 and is a member of the RING finger protein family. The E3 ubiquitin ligase MKRN1 is closely linked to tumour development, but the exact mechanism needs to be elucidated. In this study, we aimed to investigate the specific mechanism and role of MKRN1 in colorectal cancer (CRC) development. METHODS MKRN1 expression in CRC was analysed using the Cancer Cell Line Encyclopaedia and the Cancer Genome Atlas (TCGA) databases. Rectal tumour tissues were frozen to explore the MKRN1 expression in CRC and its clinical significance. The impact of MKRN1 on CRC cell proliferation and migration was observed using CCK8, colony formation, wound healing, and transwell assays. A combination of MKRN1 quantitative proteomics, ubiquitination modification omics analysis, and a string of in vitro and in vivo experiments revealed the potential mechanisms by which MKRN1 regulates CRC metastasis. RESULTS MKRN1 expression was significantly elevated in CRC tissues compared to paracancerous tissues and was positively linked with prognosis (P < 0.01). MKRN1 downregulation inhibits CRC cell proliferation, migration, and invasion. Conversely, MKRN1 overexpression promotes the proliferation, migration, and invasion of CRC cells. Mechanistically, MKRN1 induces epithelial-mesenchymal transition (EMT) in CRC cells via ubiquitination and degradation of Smad nuclear-interacting protein 1 (SNIP1). Furthermore, SNIP1 inhibits transforming growth factor-β (TGF-β) signalling, and MKRN1 promotes TGF-β signalling by degrading SNIP1 to induce EMT in CRC cells. Finally, using conditional knockout mice, intestinal lesions and metastatic liver microlesions were greatly reduced in the intestinal knockout MKRN1 group compared to that in the control group. CONCLUSIONS High MKRN1 levels promote TGF-β signalling through ubiquitination and degradation of SNIP1, thereby facilitating CRC metastasis, and supporting MKRN1 as a CRC pro-cancer factor. The MKRN1/SNIP1/TGF-β axis may be a potential therapeutic target in CRC.
Collapse
Affiliation(s)
- Yi Zhang
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
- Department of Clinical Biochemistry, School of Medical Laboratory Science, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China
| | - Qin-Shan Li
- Guizhou Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China.
- Department of Clinical Biochemistry, School of Medical Laboratory Science, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China.
| | - Hong-Lin Liu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100000, People's Republic of China
| | - Hong-Ting Tang
- Department of Clinical Biochemistry, School of Medical Laboratory Science, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China
| | - Han-Lin Yang
- Department of Clinical Biochemistry, School of Medical Laboratory Science, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China
| | - Dao-Qiu Wu
- Department of Clinical Biochemistry, School of Medical Laboratory Science, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China
| | - Yu-Ying Huang
- Department of Clinical Biochemistry, School of Medical Laboratory Science, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China
| | - Li-Cheng Li
- Clinical Medical College, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China
- Department of HematologyGuizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guizhou Province Institute of Hematology, Guizhou, Guiyang, People's Republic of China
| | - Li-Hong Liu
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China.
| | - Meng-Xing Li
- Clinical Medical College, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China.
- Department of HematologyGuizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guizhou Province Institute of Hematology, Guizhou, Guiyang, People's Republic of China.
- Department of Pathophysiology, Guizhou Medical University, Guizhou, Guiyang, 550004, People's Republic of China.
| |
Collapse
|
6
|
George M, Masamba P, Iwalokun BA, Kappo AP. Zooming into the structure-function of RING finger proteins for anti-cancer therapeutic applications. Am J Cancer Res 2023; 13:2773-2789. [PMID: 37559981 PMCID: PMC10408477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/22/2023] [Indexed: 08/11/2023] Open
Abstract
Cancer is one of the most common and widely diagnosed diseases worldwide. With an increase in prevalence and incidence, many studies in cancer biology have been looking at the role pro-cancer proteins play. One of these proteins is the Really Interesting New Gene (RING), which has been studied extensively due to its structure and functions such as apoptosis, neddylation, and its role in ubiquitination. The RING domain is a cysteine-rich domain known to bind Cysteine and Histidine residues. It also binds two zinc ions that help stabilize the protein in various patterns, often with a 'cross-brace' topology. Different RING finger proteins have been studied and found to have suitable targets for developing anti-cancer therapeutics. These identified candidate proteins include Parkin, COP1, MDM2, BARD1, BRCA-1, PIRH2, c-CBL, SIAH1, RBX1 and RNF8. Inhibiting these candidate proteins provides opportunities for shutting down pathways associated with tumour development and metastasis.
Collapse
Affiliation(s)
- Mary George
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway CampusAuckland Park, Johannesburg, South Africa
| | - Priscilla Masamba
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway CampusAuckland Park, Johannesburg, South Africa
| | - Bamidele Abiodun Iwalokun
- Department of Molecular Biology and Biotechnology, Nigerian Institute of Medical Research (NIMR)Yaba, Lagos, Nigeria
| | - Abidemi Paul Kappo
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway CampusAuckland Park, Johannesburg, South Africa
| |
Collapse
|
7
|
Kim Y, Kim EK, Chey Y, Song MJ, Jang HH. Targeted Protein Degradation: Principles and Applications of the Proteasome. Cells 2023; 12:1846. [PMID: 37508510 PMCID: PMC10378610 DOI: 10.3390/cells12141846] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The proteasome is a multi-catalytic protease complex that is involved in protein quality control via three proteolytic activities (i.e., caspase-, trypsin-, and chymotrypsin-like activities). Most cellular proteins are selectively degraded by the proteasome via ubiquitination. Moreover, the ubiquitin-proteasome system is a critical process for maintaining protein homeostasis. Here, we briefly summarize the structure of the proteasome, its regulatory mechanisms, proteins that regulate proteasome activity, and alterations to proteasome activity found in diverse diseases, chemoresistant cells, and cancer stem cells. Finally, we describe potential therapeutic modalities that use the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Yosup Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Eun-Kyung Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Yoona Chey
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Min-Jeong Song
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Ho Hee Jang
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
8
|
Han D, Wang L, Jiang S, Yang Q. The ubiquitin-proteasome system in breast cancer. Trends Mol Med 2023:S1471-4914(23)00096-5. [PMID: 37328395 DOI: 10.1016/j.molmed.2023.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Ubiquitin-proteasome system (UPS) is a selective proteolytic system that is associated with the expression or function of target proteins and participates in various physiological and pathological processes of breast cancer. Inhibitors targeting the 26S proteasome in combination with other drugs have shown promising therapeutic effects in the clinical treatment of breast cancer. Moreover, several inhibitors/stimulators targeting other UPS components are also effective in preclinical studies, but have not yet been applied in the clinical treatment of breast cancer. Therefore, it is vital to comprehensively understand the functions of ubiquitination in breast cancer and to identify potential tumor promoters or tumor suppressors among UPS family members, with the aim of developing more effective and specific inhibitors/stimulators targeting specific components of this system.
Collapse
Affiliation(s)
- Dianwen Han
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Lijuan Wang
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shan Jiang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Research Institute of Breast Cancer, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
9
|
Xi G, Cheng R, Liang L, Che N, Wang Y, Zhao N, Liang X, Shao B, Zhao X, Zhang D. High expression of RNF31 is associated with tumor immune cell infiltration and leads to poor prognosis in liver hepatocellular carcinoma. Sci Rep 2023; 13:6957. [PMID: 37117215 PMCID: PMC10147728 DOI: 10.1038/s41598-023-32692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/31/2023] [Indexed: 04/30/2023] Open
Abstract
Ring finger protein 31 (RNF31) has been found to play an important role in tumor immunity. However, the role of RNF31 in liver hepatocellular carcinoma (LIHC) has not been reported. Therefore, we investigated the expression and prognostic value of RNF31 in patients with LIHC and explored its relationship with immune cell infiltration. The Cancer Genome Atlas liver hepatocellular carcinoma (TCGA-LIHC) dataset was downloaded to analyse the impact of RNF31 on the prognosis and immune cell infiltration of LIHC. The Tumor Immune Estimation Resource (TIMER) database was used to analyse the correlation between RNF31 and tumor immune cell infiltration in LIHC. Additionally, we analysed the relationship between RNF31 and tumor necrosis factor (TNF) as well as the interferon-gamma (IFN-γ) signaling pathway. The expression of RNF31 in LIHC was significantly higher than that in normal tissues. Increased RNF31 expression was associated with decreased overall survival (OS) and relapse-free survival (RFS). An increase in RNF31 expression was closely related to the infiltration levels of immune cells (e.g., natural killer (NK) cells, CD8 + T cells, and B cells). RNF31 was also positively correlated with the expression of immune checkpoint genes in LIHC. Moreover, RNF31 may participate in TNF and IFN-γ signaling pathways. In conclusion, RNF31 is a potentially valuable prognostic biomarker in LIHC. RNF31 is also associated with immune cell infiltration in LIHC. RNF31 may be a potential target for immunotherapy of LIHC.
Collapse
Affiliation(s)
- Guifu Xi
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Runfen Cheng
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Leiting Liang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Na Che
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Yalei Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaohui Liang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Bing Shao
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
10
|
Chen Z, Ren D, Lv J, Xu Y, Xie M, He X, Shi W, Qian Q, Jing A, Ma X, Qin J, Ding Y, Geng T, Ma J, Liu W, Liu S, Ji J. The atypical ubiquitin ligase RNF31 stabilizes c-Myc via epigenetic inactivation of FBXO32 nd promotes cancer development. Cell Signal 2023; 107:110677. [PMID: 37028779 DOI: 10.1016/j.cellsig.2023.110677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/10/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023]
Abstract
RNF31, an atypical E3 ubiquitin ligase of the RING-between-RING protein family, is one of the important components of the linear ubiquitin chain complex LUBAC. It plays a carcinogenic role in a variety of cancers by promoting cell proliferation, invasion and inhibiting apoptosis. However, the specific molecular mechanism by which RNF31 exerts its cancer-promoting effects is still unclear. By analyzing the expression profile of RNF31-depleted cancer cells, we found that loss of RNF31 significantly resulted in the inactivation of the c-Myc pathway. We further showed that RNF31 played an important role in the maintenance of c-Myc protein levels in cancer cells by extending the half-life of c-Myc protein and reducing its ubiquitination. c-Myc protein levels are tightly regulated by the ubiquitin proteasome, in which the E3 ligase FBXO32 is required to mediate its ubiquitin-dependent degradation. We found that RNF31 inhibited the transcription of FBXO32 through EZH2-mediated trimethylation of histone H3K27 in the FBXO32 promoter region, leading to the stabilization and activation of c-Myc protein. Under this circumstance, the expression of FBXO32 was significantly increased in RNF31-deficient cells, promoting the degradation of c-Myc protein, inhibiting cell proliferation and invasion, increasing cell apoptosis, and ultimately blocking the progression of tumors. Consistent with these results, the reduced malignancy phenotype caused by RNF31 deficiency could be partially reversed by overexpression of c-Myc or further knockdown of FBXO32. Together, our results reveal a key association between RNF31 and epigenetic inactivation of FBXO32 in cancer cells, and suggest that RNF31 may be a promising target for cancer therapy.
Collapse
Affiliation(s)
- Zefeng Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Dexu Ren
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jinyu Lv
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yuxin Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Mengru Xie
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xingbei He
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wen Shi
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Qilan Qian
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Aixin Jing
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xinhui Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jingting Qin
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yuanyuan Ding
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Ting Geng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jinming Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wei Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Shunfang Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan 430030, P.R. China.
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
11
|
Zhang J, Tu H, Zheng Z, Zhao X, Lin X. RNF31 promotes tumorigenesis via inhibiting RIPK1 kinase-dependent apoptosis. Oncogene 2023; 42:1585-1596. [PMID: 36997719 DOI: 10.1038/s41388-023-02669-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023]
Abstract
It is well established that interferon (IFN) and tumor necrosis factor (TNF) could synergistically promote antitumor toxicity and avoid resistance of antigen-negative tumors during cancer immunotherapy. The linear ubiquitin chain assembly complex (LUBAC) has been widely known to regulate receptor-interacting protein kinase-1(RIPK1) kinase activity and TNF-mediated cell death during inflammation and embryogenesis. However, whether LUBAC and RIPK1 kinase activity in tumor microenvironment could regulate antitumor immunity are still not very clear. Here, we demonstrated a cancer cell-intrinsic role of LUBAC complex in tumor microenvironment to promote tumorigenesis. Lacking LUBAC component RNF31 in B16 melanoma cells but not immune cells including macrophages or dendritic cells greatly impaired tumor growth by increasing intratumoral CD8+ T cells infiltration. Mechanistically, we found that tumor cells without RNF31 shown severe apoptosis-mediated cell death caused by TNFα/IFNγ in the tumor microenvironment. Most importantly, we found that RNF31 could limit RIPK1 kinase activity and further prevent tumor cell death in a transcription-independent manner, suggesting a crucial role of RIPK1 kinase activity in tumorigenesis. Together, our results demonstrate an essential role of RNF31 and RIPK1 kinase activity in tumorigenesis and imply that RNF31 inhibition could be harnessed to enhance antitumor toxicity during tumor immunotherapy.
Collapse
Affiliation(s)
- Jie Zhang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Hailin Tu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Zheyu Zheng
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xueqiang Zhao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xin Lin
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China.
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China.
| |
Collapse
|
12
|
Yang H, Xue M, Su P, Zhou Y, Li X, Li Z, Xia Y, Zhang C, Fu M, Zheng X, Luo G, Wei T, Wang X, Ding Y, Zhu J, Zhuang T. RNF31 represses cell progression and immune evasion via YAP/PD-L1 suppression in triple negative breast Cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:364. [PMID: 36581998 PMCID: PMC9801641 DOI: 10.1186/s13046-022-02576-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/19/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Recently genome-based studies revealed that the abnormality of Hippo signaling is pervasive in TNBC and played important role in cancer progression. RING finger protein 31 (RNF31) comes to RING family E3 ubiquitin ligase. Our previously published studies have revealed RNF31 is elevated in ER positive breast cancer via activating estrogen signaling and suppressing P53 pathway. METHODS We used several TNBC cell lines and xenograft models and performed immuno-blots, QPCR, in vivo studies to investigate the function of RNF31 in TNBC progression. RESULT Here, we demonstrate that RNF31 plays tumor suppressive function in triple negative breast cancer (TNBC). RNF31 depletion increased TNBC cell proliferation and migration in vitro and in vitro. RNF31 depletion in TNBC coupled with global genomic expression profiling indicated Hippo signaling could be the potential target for RNF31 to exert its function. Further data showed that RNF31 depletion could increase the level of YAP protein, and Hippo signaling target genes expression in several TNBC cell lines, while clinical data illustrated that RNF31 expression correlated with longer relapse-free survival in TNBC patients and reversely correlated with YAP protein level. The molecular biology assays implicated that RNF31 could associate with YAP protein, facilitate YAP poly-ubiquitination and degradation at YAP K76 sites. Interestingly, RNF31 could also repress PDL1 expression and sensitive TNBC immunotherapy via inhibiting Hippo/YAP/PDL1 axis. CONCLUSIONS Our study revealed the multi-faced function of RNF31 in different subtypes of breast malignancies, while activation RNF31 could be a plausible strategy for TNBC therapeutics.
Collapse
Affiliation(s)
- Huijie Yang
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Min Xue
- grid.440265.10000 0004 6761 3768Molecular Biology Laboratory, First People’s Hospital of Shangqiu, Shangqiu, City, 476000 Henan Province People’s Republic of China
| | - Peng Su
- Department of Pathology, Shandong University Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, Shandong Province People’s Republic of China
| | - Yan Zhou
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Xin Li
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Zhongbo Li
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Yan Xia
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Chenmiao Zhang
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Mingxi Fu
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Xiuxia Zheng
- grid.440265.10000 0004 6761 3768Molecular Biology Laboratory, First People’s Hospital of Shangqiu, Shangqiu, City, 476000 Henan Province People’s Republic of China
| | - Guosheng Luo
- grid.412990.70000 0004 1808 322XThe Affiliated people’s Hospital of Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Tian Wei
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Xinxing Wang
- grid.412633.10000 0004 1799 0733Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052 People’s Republic of China
| | - Yinlu Ding
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Jian Zhu
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Ting Zhuang
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China ,grid.412990.70000 0004 1808 322XThe Affiliated people’s Hospital of Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| |
Collapse
|
13
|
Chen X, Ye Q, Zhao W, Chi X, Xie C, Wang X. RBCK1 promotes hepatocellular carcinoma metastasis and growth by stabilizing RNF31. Cell Death Discov 2022; 8:334. [PMID: 35869046 PMCID: PMC9307510 DOI: 10.1038/s41420-022-01126-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 12/24/2022] Open
Abstract
AbstractRNF31 (HOIP), RBCK1 (HOIL-1L), and SHARPIN are subunits of the linear ubiquitin chain assembly complex. Their function and specific molecular mechanisms in hepatocellular carcinoma (HCC) have not been reported previously. Here, we investigated the role of RNF31 and RBCK1 in HCC. We showed that RNF31 and RBCK1 were overexpressed in HCC and that upregulation of RNF31 and RBCK1 indicated poor clinical outcomes in patients with HCC. RNF31 overexpression was significantly associated with more satellite foci and vascular invasion in patients with HCC. Additionally, RBCK1 expression correlated positively with RNF31 expression in HCC tissues. Functionally, RBCK1 and RNF31 promote the metastasis and growth of HCC cells. Moreover, the RNF31 inhibitor gliotoxin inhibited the malignant behavior of HCC cells. Mechanistically, RBCK1 interacted with RNF31 and repressed its ubiquitination and proteasomal degradation. In summary, the present study revealed an oncogenic role and regulatory relationship between RBCK1 and RNF31 in facilitating proliferation and metastasis in HCC, suggesting that they are potential prognostic markers and therapeutic targets for HCC.
Collapse
|
14
|
Liu J, Wang Q, Kang Y, Xu S, Pang D. Unconventional protein post-translational modifications: the helmsmen in breast cancer. Cell Biosci 2022; 12:22. [PMID: 35216622 PMCID: PMC8881842 DOI: 10.1186/s13578-022-00756-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
AbstractBreast cancer is the most prevalent malignant tumor and a leading cause of mortality among females worldwide. The tumorigenesis and progression of breast cancer involve complex pathophysiological processes, which may be mediated by post-translational modifications (PTMs) of proteins, stimulated by various genes and signaling pathways. Studies into PTMs have long been dominated by the investigation of protein phosphorylation and histone epigenetic modifications. However, with great advances in proteomic techniques, several other PTMs, such as acetylation, glycosylation, sumoylation, methylation, ubiquitination, citrullination, and palmitoylation have been confirmed in breast cancer. Nevertheless, the mechanisms, effects, and inhibitors of these unconventional PTMs (particularly, the non-histone modifications other than phosphorylation) received comparatively little attention. Therefore, in this review, we illustrate the functions of these PTMs and highlight their impact on the oncogenesis and progression of breast cancer. Identification of novel potential therapeutic drugs targeting PTMs and development of biological markers for the detection of breast cancer would be significantly valuable for the efficient selection of therapeutic regimens and prediction of disease prognosis in patients with breast cancer.
Collapse
|
15
|
Guo Y, He J, Zhang H, Chen R, Li L, Liu X, Huang C, Qiang Z, Zhou Z, Wang Y, Huang J, Zhao X, Zheng J, Chen GQ, Yu J. Linear ubiquitination of PTEN impairs its function to promote prostate cancer progression. Oncogene 2022; 41:4877-4892. [PMID: 36192478 DOI: 10.1038/s41388-022-02485-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022]
Abstract
PTEN is frequently mutated in human cancers, which leads to the excessive activation of PI3K/AKT signaling and thus promotes tumorigenesis and drug resistance. Met1-linked ubiquitination (M1-Ubi) is also involved in cancer progression, but the mechanism is poorly defined. Here we find that HOIP, one important component of linear ubiquitin chain assembly complex (LUBAC), promotes prostate cancer (PCa) progression by enhancing AKT signaling in a PTEN-dependent manner. Mechanistically, PTEN is modified by M1-Ubi at two sites K144 and K197, which significantly inhibits PTEN phosphatase activity and thus accelerates PCa progression. More importantly, we identify that the high-frequency mutants PTENR173H and PTENR173C in PCa patients showed the enhanced level of M1-Ubi, which impairs PTEN function in inhibition of AKT phosphorylation and cell growth. We also find that HOIP depletion sensitizes PCa cells to therapeutic agents BKM120 and Enzalutamide. Furthermore, the clinical data analyses confirm that HOIP is upregulated and positively correlated with AKT activation in PCa patient specimen, which may promote PCa progression and increase the risk of PCa biochemical relapse. Together, our study reveals a key role of PTEN M1-Ubi in regulation of AKT activation and PCa progression, which may propose a new strategy for PCa therapy.
Collapse
Affiliation(s)
- Yanmin Guo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianfeng He
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hailong Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ran Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lian Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaojia Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Caihu Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhe Qiang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zihan Zhou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanli Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xian Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junke Zheng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guo-Qiang Chen
- State Key Laboratory of Oncogenes and Related Genes, Ren-Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
16
|
Jiao F, Tan Z, Yu Z, Zhou B, Meng L, Shi X. The phytochemical and pharmacological profile of taraxasterol. Front Pharmacol 2022; 13:927365. [PMID: 35991893 PMCID: PMC9386448 DOI: 10.3389/fphar.2022.927365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
Taraxasterol is one of the bioactive triterpenoids found in dandelion, a member of the family Asteraceae. In the animal or cellular models of several ailments, including liver damage, gastritis, colitis, arthritis, pneumonia, tumors, and immune system diseases, taraxasterol has been shown to have significant preventive and therapeutic effects. This review aims to evaluate the current state of research and provide an overview of the possible applications of taraxasterol in various diseases. The reported phytochemical properties and pharmacological actions of taraxasterol, including anti-inflammatory, anti-oxidative, and anti-carcinogenic properties, and its potential molecular mechanisms in developing these diseases are highlighted. Finally, we further explored whether taraxasterol has protective effects on neuronal death in neurodegenerative diseases. In addition, more animal and clinical studies are also required on the metabolism, bioavailability, and safety of taraxasterol to support its applications in pharmaceuticals and medicine.
Collapse
Affiliation(s)
- Fengjuan Jiao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
- *Correspondence: Fengjuan Jiao,
| | - Zengyue Tan
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Zhonghua Yu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Bojie Zhou
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Lingyan Meng
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Xinyue Shi
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| |
Collapse
|
17
|
Li X, Niu Z, Sun C, Zhuo S, Yang H, Yang X, Liu Y, Yan C, Li Z, Cao Q, Ji G, Ding Y, Zhuang T, Zhu J. Regulation of P53 signaling in breast cancer by the E3 ubiquitin ligase RNF187. Cell Death Dis 2022; 13:149. [PMID: 35165289 PMCID: PMC8844070 DOI: 10.1038/s41419-022-04604-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 01/19/2022] [Accepted: 02/01/2022] [Indexed: 01/15/2023]
Abstract
The tumor suppressor P53 plays critical role in preventing cancer. P53 is rarely mutated and remains functional in luminal-type breast cancer(1). According to current knowledge, wild-type P53 function is tightly controlled by posttranslational modifications, such as ubiquitination. Several ubiquitin ligases have been shown to regulate P53 ubiquitination and protein stability. Here, we report that RNF187, a RING family ubiquitin ligase, facilitates breast cancer growth and inhibits apoptosis by modulating P53 signaling. RNF187 expression was elevated in breast cancer and correlated with breast cancer survival only in the P53 wild-type groups. Bioinformatic analysis showed that the expression of RNF187 was negatively correlated with the expression of P53 target genes, such as IGFBP3 and FAS, in breast cancer. RNF187 depletion inhibited breast cancer growth and facilitated cell death. RNA sequencing analysis indicated that RNF187 could be an important modulator of P53 signaling. Further experiments showed that RNF187 interacts with P53 and promotes its degradation by facilitating its polyubiquitination in breast cancer cells. Interestingly, the in vitro ubiquitin assay showed that RNF187 can directly ubiquitinate P53 in a manner independent of MDM2. These findings reveal a novel direct P53 regulator and a potential therapeutic target for breast cancer.
Collapse
|
18
|
Freeman AJ, Vervoort SJ, Michie J, Ramsbottom KM, Silke J, Kearney CJ, Oliaro J. HOIP limits anti-tumor immunity by protecting against combined TNF and IFN-gamma-induced apoptosis. EMBO Rep 2021; 22:e53391. [PMID: 34467615 DOI: 10.15252/embr.202153391] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
The success of cancer immunotherapy is limited to a subset of patients, highlighting the need to identify the processes by which tumors evade immunity. Using CRISPR/Cas9 screening, we reveal that melanoma cells lacking HOIP, the catalytic subunit of LUBAC, are highly susceptible to both NK and CD8+ T-cell-mediated killing. We demonstrate that HOIP-deficient tumor cells exhibit increased sensitivity to the combined effect of the inflammatory cytokines, TNF and IFN-γ, released by NK and CD8+ T cells upon target recognition. Both genetic deletion and pharmacological inhibition of HOIP augment tumor cell sensitivity to combined TNF and IFN-γ. Together, we unveil a protective regulatory axis, involving HOIP, which limits a transcription-dependent form of cell death that engages both intrinsic and extrinsic apoptotic machinery upon exposure to TNF and IFN-γ. Our findings highlight HOIP inhibition as a potential strategy to harness and enhance the killing capacity of TNF and IFN-γ during immunotherapy.
Collapse
Affiliation(s)
- Andrew J Freeman
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia
| | - Stephin J Vervoort
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia.,Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - Jessica Michie
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia
| | - Kelly M Ramsbottom
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - John Silke
- Inflammation Department, Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Vic., Australia
| | - Conor J Kearney
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia.,Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - Jane Oliaro
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic., Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Vic., Australia
| |
Collapse
|
19
|
Niu Z, Li X, Dong S, Gao J, Huang Q, Yang H, Qian H, Zhuo S, Zhuang T, Zhu J, Ding Y, Xu W. The E3 Ubiquitin Ligase HOIP inhibits Cancer Cell Apoptosis via modulating PTEN stability. J Cancer 2021; 12:6553-6562. [PMID: 34659546 PMCID: PMC8489130 DOI: 10.7150/jca.61996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/30/2021] [Indexed: 11/22/2022] Open
Abstract
Chemotherapy is widely used in a variety of solid tumors, such as lung cancer, gastric cancer and breast cancer. The genotoxic drugs, such as cisplatin, suppress cancer progression either by inhibition cell proliferation or facilitating apoptosis. However, the chemotherapy resistance remains an urgent challenge in cancer therapy, especially in advanced stages. Several studies showed that the activation of pro-survival pathways, such as PI3K-AKT, participated in mediating chemotherapy resistance. The insights into the molecular mechanisms for underlying chemotherapy resistance are of great importance to improve cancer patient survival in advanced stages. The HOIP protein belongs to the RING family E3 ubiquitin ligases and modulates several atypical ubiquitination processes in cellular signaling. Previous studies showed that HOIP might be an important effector in modulating cancer cell death under genotoxic drugs. Here, we report that HOIP associates with PTEN and facilitates PTEN degradation in cancer cells. Depletion of HOIP causes cell cycle arrest and apoptosis, which effects could be rescued by PTEN silencing. Besides, the survival data from public available database show that HOIP expression correlates with poor survival in several types of chemotherapy-treated cancer patients. In conclusion, our study establishes a novel mechanism by which HOIP modulates PTEN stability and facilitates chemotherapy resistance in malignancies.
Collapse
Affiliation(s)
- Zhiguo Niu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212000, China.,Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Xin Li
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Shuxiao Dong
- Department of Gastroenterology surgery, Shandong Provincial Third Hospital, Jinan, 250000, China
| | - Jianhui Gao
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Qingsong Huang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Huijie Yang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Shu Zhuo
- Signet Therapeutics Inc, Shenzhen, China. Research Institute of Tsinghua University in Shenzhen, Shenzhen, 518000, China
| | - Ting Zhuang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Jian Zhu
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China.,Department of general surgery, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Yinlu Ding
- Department of general surgery, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| |
Collapse
|
20
|
Zhang J, Jiang X, Yin J, Dou S, Xie X, Liu T, Wang Y, Wang S, Zhou X, Zhang D, Jiang H. RNF141 interacts with KRAS to promote colorectal cancer progression. Oncogene 2021; 40:5829-5842. [PMID: 34345014 PMCID: PMC8484013 DOI: 10.1038/s41388-021-01877-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/22/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023]
Abstract
RING finger proteins (RNFs) play a critical role in cancer initiation and progression. RNF141 is a member of RNFs family; however, its clinical significance, roles, and mechanism in colorectal cancer (CRC) remain poorly understood. Here, we examined the expression of RNF141 in 64 pairs of CRC and adjacent normal tissues by real-time PCR, Western blot, and immunohistochemical analysis. We found that there was more expression of RNF141 in CRC tissue compared with its adjacent normal tissue and high RNF141 expression associated with T stage. In vivo and in vitro functional experiments were conducted and revealed the oncogenic role of RNF141 in CRC. RNF141 knockdown suppressed proliferation, arrested the cell cycle in the G1 phase, inhibited migration, invasion and HUVEC tube formation but promoted apoptosis, whereas RNF141 overexpression exerted the opposite effects in CRC cells. The subcutaneous xenograft models showed that RNF141 knockdown reduced tumor growth, but its overexpression promoted tumor growth. Mechanistically, liquid chromatography-tandem mass spectrometry indicated RNF141 interacted with KRAS, which was confirmed by Co-immunoprecipitation, Immunofluorescence assay. Further analysis with bimolecular fluorescence complementation (BiFC) and Glutathione-S-transferase (GST) pull-down assays showed that RNF141 could directly bind to KRAS. Importantly, the upregulation of RNF141 increased GTP-bound KRAS, but its knockdown resulted in a reduction accordingly. Next, we demonstrated that RNF141 induced KRAS activation via increasing its enrichment on the plasma membrane not altering total KRAS expression, which was facilitated by the interaction with LYPLA1. Moreover, KRAS silencing partially abolished the effect of RNF141 on cell proliferation and apoptosis. In addition, our findings presented that RNF141 functioned as an oncogene by upregulating KRAS activity in a manner of promoting KRAS enrichment on the plasma membrane in CRC.
Collapse
Affiliation(s)
- Jiuna Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
- Department of Gastroenterology, The Affiliated Hospital of Hebei Engineering University, Handan, P. R. China
| | - Xiaoyu Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Jie Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Shiying Dou
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Ting Liu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Yijun Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Shuling Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Xue Zhou
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Dongxuan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China
| | - Huiqing Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang, P. R. China.
| |
Collapse
|
21
|
Cheng Y, Hu Y, Wang H, Zhao Z, Jiang X, Zhang Y, Zhang J, Tong Y, Qiu X. Ring finger protein 19A is overexpressed in non-small cell lung cancer and mediates p53 ubiquitin-degradation to promote cancer growth. J Cell Mol Med 2021; 25:7796-7808. [PMID: 34184814 PMCID: PMC8358885 DOI: 10.1111/jcmm.16674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/19/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
The expression pattern, biological functions and the related mechanisms of the ring finger protein 19A (RNF19A) in non-small cell lung cancer (NSCLC) remain poorly understood. This study aimed to explore the role of RNF19A, as well as the underlying potential mechanism, in the development of NSCLC. Here, we found that RNF19A was overexpressed in NSCLC tissues, and RNF19A expression in NSCLC tissue samples was associated with NSCLC carcinogenesis and poor outcome. RNF19A promoted the proliferation of NSCLC cells and inhibited apoptosis. RNF19A reduced p53, p21 and BAX expression and induced Cyclin D1, CDK4, CDK6 and BCL2 expression. The inhibitory effect of RNF19A knockdown on proliferation was partially rescued by p53 silencing. RNF19A interacted with p53, shortened p53 half-life and mediated p53 ubiquitin-degradation. Collectively, we suggest that RNF19A plays a critical oncogenic role in lung carcinogenesis by disrupting the function of p53. RNF19A may serve as a new biomarker and/or target for NSCLC management.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Pathology, Cancer Research Laboratory, Chengde Medical College, Chengde, China
| | - Yujiao Hu
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Huanxi Wang
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Zhi Zhao
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Pathology, Zhengzhou Yihe Hospital Affiliated to Henan University, Zhengzhou, China
| | - Xizi Jiang
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yao Zhang
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jiameng Zhang
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yue Tong
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xueshan Qiu
- Department of Pathology, College of Basic Medical Sciences and First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
22
|
Tang CT, Yang J, Liu ZD, Chen Y, Zeng C. Taraxasterol acetate targets RNF31 to inhibit RNF31/p53 axis-driven cell proliferation in colorectal cancer. Cell Death Discov 2021; 7:66. [PMID: 33824292 PMCID: PMC8024285 DOI: 10.1038/s41420-021-00449-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/22/2021] [Accepted: 03/08/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. Several studies have suggested that taraxasterol acetate (TA) can inhibit the growth of tumor cells. However, to date, it remains unclear how TA inhibits cell growth and how RNF31 functions as an oncogene. We examined the expression of RNF31 in CRC tissue samples via immunohistochemistry and elucidated the function of RNF31 in CRC cells by constructing a cell model with RNF31 depletion. A cycloheximide (CHX)-chase analysis and immunofluorescence assays were conducted to demonstrate that TA can promote RNF31 degradation by activating autophagy. We used the PharmMapper website to predict targets of TA and identified RNF31. CHX-chase experiments showed that TA could facilitate RNF31 degradation, which was inhibited by the administration of chloroquine. Immunofluorescence assays showed that RNF31 protein was colocalized with LC3I/II and p62, suggesting that TA promoted RNF31 degradation by activating autophagy. We also found that CRC patients with RNF31 overexpression had poorer survival than those with low RNF31 expression. The results of the CHX-chase experiment showed that depletion of RNF31 alleviated p53 degradation, which was inhibited by MG132. A series of co-immunoprecipitation (Co-IP) assays revealed that RNF31 interacts with p53 and promotes p53 ubiquitination and degradation. A Co-IP assay performed with a truncated RNF31 plasmid showed that the PUB domain interacts with p53. Moreover, the PUB domain is the key structure in the induction of p53 ubiquitination. Our findings reveal a key role of RNF31 in CRC cell growth and indicate a mechanism through which TA inhibits cell growth.
Collapse
Affiliation(s)
- Chao-Tao Tang
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Yang
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zi-De Liu
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Youxiang Chen
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Chunyan Zeng
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
23
|
Yang H, Lv X, Li X, Mao L, Niu Z, Wang T, Zhuang T, Huang Q. ZNF213 Facilitates ER Alpha Signaling in Breast Cancer Cells. Front Oncol 2021; 11:638751. [PMID: 33777799 PMCID: PMC7987952 DOI: 10.3389/fonc.2021.638751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
Background Breast cancer is the most common women malignancy worldwide, while estrogen receptor alpha positive type accounts for two third of all breast cancers. Although ER alpha positive breast cancer could be effectively controlled by endocrine therapy, more than half of the cases could develop endocrine resistance, making it an important clinical issue in breast cancer treatment. Thus, decoding the detailed mechanism, which controls ER alpha signaling activation and ER alpha protein stability, is of great importance for the improvement of breast cancer therapy. Several zinc finger proteins were shown to mediate the ubiquitination process and modulate protein stability. Thus, we further explore the function of Zinc finger protein 213 on ER alpha protein stability and tamoxifen resistance. Methods CCK8 and Edu assay was used to measure cell proliferation. RNA sequence was performed by Ingenuity pathway analysis. The ER alpha signaling activities were measured with luciferase assay, real-time quantitative PCR, and western blotting. Protein stability assay and ubiquitin assay were used to determine ER alpha protein degradation and ubiquitination. The immuno-precipitation was utilized to determine ER alpha and ZNF213 interaction. The ubiquitin-based immuno-precipitation assay was sued to detect specific ubiquitination manner on ER alpha. Results We identified ZNF213 as a novel zinc finger protein, which modulated ER alpha protein. ZNF213 expression correlated with poor outcome in endocrine treated patients. ZNF213 depletion inhibited ER alpha signaling and proliferation in breast cancer cells. Further mechanistic studies showed ZNF213 located in cytosol and nuclear, which modulated ER alpha stability via inhibiting ER alpha K48-linked ubiquitination. Conclusions Our study reveals an interesting post-translational mechanism between ER alpha and ZNF213 in breast cancer. Targeting ZNF213 could be an appealing strategy for ER alpha positive breast cancer.
Collapse
Affiliation(s)
- Huijie Yang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Xinxiang Key Laboratory of Tumor Migration, Invasion and Precision Medicine, Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Xulei Lv
- Department of Anesthesiology, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xin Li
- Xinxiang Key Laboratory of Tumor Migration, Invasion and Precision Medicine, Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Lanzhi Mao
- Xinxiang Key Laboratory of Tumor Migration, Invasion and Precision Medicine, Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Zhiguo Niu
- Xinxiang Key Laboratory of Tumor Migration, Invasion and Precision Medicine, Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Ting Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ting Zhuang
- Xinxiang Key Laboratory of Tumor Migration, Invasion and Precision Medicine, Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Qingsong Huang
- Xinxiang Key Laboratory of Tumor Migration, Invasion and Precision Medicine, Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
24
|
Prognostic significance of low expression of B-cell translocation gene 1 (BTG 1) in skin squamous cell carcinoma. J Biosci 2020. [DOI: 10.1007/s12038-020-00032-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
Quan Y, Liang F, Wu D, Yao X, Hu Z, Zhu Y, Chen Y, Wu A, Tang D, Huang B, Xu R, Lyu Z, Yan Q, Luo L, Ning Z, Li Y, Xiong J. Blood Cell DNA Methylation of Aging-Related Ubiquitination Gene DZIP3 Can Predict the Onset of Early Stage Colorectal Cancer. Front Oncol 2020; 10:544330. [PMID: 33330022 PMCID: PMC7729090 DOI: 10.3389/fonc.2020.544330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 10/28/2020] [Indexed: 12/27/2022] Open
Abstract
There is a body of evidence that the aging immune system is linked to cancer. In this study, with aging- and immune-related DNA methylation data, we investigated the DNA methylation regulation changes in promoters with other regions of genes during aging and their association with the immune-cell proportion in the circulating whole blood of individuals. The analyses for aging- and CD4+ T cell proportion-derived differential genes showed that ubiquitination plays an important role in the aging immune system and tumorigenesis. Therefore, starting from a set of pre-annotated ubiquitination genes, we found that among the differentially ubiquitinated genes, DZIP3, an E3 ubiquitin ligase with no reports on its function in immune cells and tumorigenesis, was significantly associated with both aging (P-value = 3.86e-06) and CD4+ T cell proportion (P-value = 1.97e-05) in circulating blood. By collecting a cohort of 100 colon cancer patients and 50 healthy individuals, we validated that the 1st exon DNA methylation of DZIP3 could predict the onset of early stage (AUC = 0.833, OR = 8.82) and all pTNM stages of colorectal cancer (AUC = 0.782, OR = 5.70). Thus, the epigenetically regulated ubiquitination machine plays an important role in immune aging and tumorigenesis.
Collapse
Affiliation(s)
- Yuan Quan
- School of Computer Science and Technology, Harbin Institute of Technology Shenzhen Graduate School, Shenzhen, China
- Lab of Epigenetics and Advanced Health Technology, Space Science and Technology Institute (Shenzhen), Shenzhen, China
| | - Fengji Liang
- Lab of Epigenetics and Advanced Health Technology, Space Science and Technology Institute (Shenzhen), Shenzhen, China
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Deqing Wu
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xueqing Yao
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhihuang Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuexing Zhu
- Lab of Epigenetics and Advanced Health Technology, Space Science and Technology Institute (Shenzhen), Shenzhen, China
| | - Ying Chen
- Lab of Epigenetics and Advanced Health Technology, Space Science and Technology Institute (Shenzhen), Shenzhen, China
| | - Andong Wu
- Lab of Epigenetics and Advanced Health Technology, Space Science and Technology Institute (Shenzhen), Shenzhen, China
| | - Danian Tang
- Gastro-Intestinal Surgery Department, Beijing Hospital, Beijing, China
| | - Bingyang Huang
- Department of Cardiothoracic Surgery, Strategic Support Force Medical Center of PLA. No. 9, Beijing, China
| | - Ruifeng Xu
- School of Computer Science and Technology, Harbin Institute of Technology Shenzhen Graduate School, Shenzhen, China
| | - Zejian Lyu
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qian Yan
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lang Luo
- Shenzhen Taikontek Health Technology Co., Ltd, Shenzhen, China
| | - Zhengzhi Ning
- Shenzhen Taikontek Health Technology Co., Ltd, Shenzhen, China
| | - Yong Li
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jianghui Xiong
- Lab of Epigenetics and Advanced Health Technology, Space Science and Technology Institute (Shenzhen), Shenzhen, China
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
26
|
Abstract
BACKGROUND Beast cancer is the most common women cancer worldwide, while two third of them are ER alpha positive breast cancer. Among the ER alpha positive breast cancer, about 80% are P53 wild type, indicating the potential tumor suppression role in ER alpha positive breast cancer. Since P53 is an important safeguard to inhibit cell malignant transformation, reactivating P53 signaling could a plausible approach to treat breast cancer. METHODS TRIM3 protein levels were measured by western blot, while the P53 classical target genes were measured by real-time PCR. WST1 assay were used to measure cell proliferation, while cleaved caspase-3 was used to detect cell apoptosis. Protein stability and ubiquitin assay were used to detect the P53 protein ubiquitin and stability. The immuno-precipitation assays were used to detect the protein interactions. Immuno-staining was used to detect the protein localization of P53 and TRIM3, while the ubiquitin-based immuno-precipitation assays were used to detect the specific ubiquitination manner of P53. RESULTS In our study, we identified TRIM3 as an endogenous inhibitor for P53 signaling. TRIM3 depletion inhibited breast cancer cell proliferation and promoted apoptosis. In addition, TRIM3 depletion increased P53 protein level in breast cancer cell. Further investigation showed that TRIM3 could associate with P53 and promote P53 K48-linked ubiquitination and degradation. CONCLUSION Our study identified a novel post-translational modification mechanism between TRIM3 and P53. TRIM3 depletion or blockage could be a promising strategy to rescue P53 signaling and inhibit breast cancer progression.
Collapse
|
27
|
Pu Q, Lv YR, Dong K, Geng WW, Gao HD. Tumor suppressor OTUD3 induces growth inhibition and apoptosis by directly deubiquitinating and stabilizing p53 in invasive breast carcinoma cells. BMC Cancer 2020; 20:583. [PMID: 32571254 PMCID: PMC7310228 DOI: 10.1186/s12885-020-07069-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 06/15/2020] [Indexed: 12/20/2022] Open
Abstract
Background P53 pathway inactivation plays an important role in the process of breast cancer tumorigenesis. Post-translational protein modification abnormalities have been confirmed to be an important mechanism underlying inactivation of p53. Numerous deubiquitinating enzymes are aberrantly expressed in breast cancer, and a few deubiquitination enzymes can deubiquitinate and stabilize p53. Here, we report that ovarian tumor (OTU) deubiquitinase 3 (OTUD3) is a deubiquitylase of p53 in breast carcinoma (BC). Methods Correlations between the mRNA expression levels of OTUD3, TP53 and PTEN and the prognosis of BC were assessed with the Kaplan-Meier Plotter tool. OTUD3 protein expression in 80 pairs of specimens in our cohort was examined by immunohistochemistry and western blotting. The relationship among OTUD3, p53, and p21 proteins was analyzed. Half-life analysis and ubiquitylation assay were performed to elucidate the molecular mechanism by which OTUD3 stabilizes p53. The interaction between OTUD3 and p53 in BC cells was verified by a co-immunoprecipitation assay and GST pulldown experiments. MTS assay for proliferation detection, detection of apoptosis induced by cisplatin and colony formation assay were employed to investigate the functional effects of OTUD3 on breast cancer cells. Results OTUD3 downregulation is correlated with a poor prognosis in BC patients. OTUD3 expression is decreased in breast cancer tissues and not associated with the histological grade. OTUD3 also inhibits cell proliferation and clone formation and increases the sensitivity of BC cells to apoptosis induced by chemotherapy drugs. Reduced OTUD3 expression accompanied by decreased p53 abundance is correlated with human breast cancer progression. Ectopic expression of wild-type OTUD3, but not its catalytically inactive mutant, stabilizes and activates p53. Mechanistically, OTUD3 interacts directly with p53 through the amino-terminal OTU region. Finally, OTUD3 protects p53 from murine double minute 2 (Mdm2)-mediated ubiquitination and degradation, enabling the deubiquitination of p53 in BC cells. Conclusions In summary, we found that OTUD3 may be a potential therapeutic target for restoring p53 function in breast cancer cells and suggest that the OTUD3-p53 signaling axis may play a critical role in tumor suppression.
Collapse
Affiliation(s)
- Qian Pu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China.,Department of General Surgery, Qilu Hospital (Qingdao) of Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, P.R. China
| | - Yan-Rong Lv
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Ke Dong
- Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Wen-Wen Geng
- Department of General Surgery, Qilu Hospital (Qingdao) of Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, P.R. China
| | - Hai-Dong Gao
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China. .,Department of General Surgery, Qilu Hospital (Qingdao) of Shandong University, 758 Hefei Road, Qingdao, Shandong, 266035, P.R. China.
| |
Collapse
|
28
|
Wang P, Dai X, Jiang W, Li Y, Wei W. RBR E3 ubiquitin ligases in tumorigenesis. Semin Cancer Biol 2020; 67:131-144. [PMID: 32442483 DOI: 10.1016/j.semcancer.2020.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
RING-in-between-RING (RBR) E3 ligases are one class of E3 ligases that is characterized by the unique RING-HECT hybrid mechanism to function with E2s to transfer ubiquitin to target proteins for degradation. Emerging evidence has demonstrated that RBR E3 ligases play essential roles in neurodegenerative diseases, infection, inflammation and cancer. Accumulated evidence has revealed that RBR E3 ligases exert their biological functions in various types of cancers by modulating the degradation of tumor promoters or suppressors. Hence, we summarize the differential functions of RBR E3 ligases in a variety of human cancers. In general, ARIH1, RNF14, RNF31, RNF144B, RNF216, and RBCK1 exhibit primarily oncogenic roles, whereas ARIH2, PARC and PARK2 mainly have tumor suppressive functions. Moreover, the underlying mechanisms by which different RBR E3 ligases are involved in tumorigenesis and progression are also described. We discuss the further investigation is required to comprehensively understand the critical role of RBR E3 ligases in carcinogenesis. We hope our review can stimulate the researchers to deeper explore the mechanism of RBR E3 ligases-mediated carcinogenesis and to develop useful inhibitors of these oncogenic E3 ligases for cancer therapy.
Collapse
Affiliation(s)
- Peter Wang
- School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, China
| | - Xiaoming Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA, USA
| | - Wenxiao Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Yuyun Li
- School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, China.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA, USA.
| |
Collapse
|
29
|
Gong L, Xiao M, He D, Hu Y, Zhu Y, Xiang L, Bao Y, Liu X, Zeng Q, Liu J, Zhou M, Zhou Y, Cheng Y, Zhang Y, Deng L, Zhu R, Lan H, Cao K. WDHD1 Leads to Cisplatin Resistance by Promoting MAPRE2 Ubiquitination in Lung Adenocarcinoma. Front Oncol 2020; 10:461. [PMID: 32426268 PMCID: PMC7212426 DOI: 10.3389/fonc.2020.00461] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/13/2020] [Indexed: 12/23/2022] Open
Abstract
Ubiquitin ligases have been shown to regulate drug sensitivity. This study aimed to explore the role of the ubiquitin ligase WD repeat and HMG-box DNA binding protein 1 (WDHD1) in regulating cisplatin sensitivity in lung adenocarcinoma (LUAD). A quantitative analysis of the global proteome identified differential protein expression between LUAD A549 cells and the cisplatin-resistant strain A549/DDP. Public databases revealed the relationship between ubiquitin ligase expression and the prognosis of patients with LUAD. Quantitative real-time polymerase chain reaction and Western blotting were used to estimate the WDHD1 expression levels. Analysis of public databases predicted the substrate of WDHD1. Western blotting detected the effect of WDHD1 on microtubule-associated protein RP/EB family member 2 (MAPRE2) and DSTN. Functional analysis of MAPRE2 verified the interaction between WDHD1 and MAPRE2, as well as the interacting sites by methyl-thiazolyl-tetrazolium assay and flow cytometry, immunoprecipitation, protein stability, and immunofluorescence. Cell and animal experiments confirmed the effect of WDHD1 and MAPRE2 on cisplatin sensitivity in LUAD. Clinical data evaluated the impact of WDHD1 expression level on cisplatin sensitivity. Quantitative analysis of the global proteome revealed ubiquitin-dependent protein catabolism to be more active in A549/DDP cells than in A549 cells. WDHD1 expression was higher in A549/DDP cells than in A549 cells, and knocking out WDHD1 increased the sensitivity of A549/DDP cells to cisplatin. WDHD1 overexpression negatively correlated with the overall survival of LUAD patients. We observed that MAPRE2 was upregulated when WDHD1 was knocked out. A MAPRE2 knockout in A549 cells resulted in increased cell viability while decreasing apoptosis when the A549 cells exposed to cisplatin. WDHD1 and MAPRE2 were found to interact in the nucleus, and WDHD1 promoted the ubiquitination of MAPRE2. Following cisplatin exposure, the WDHD1 and MAPRE2 knockout groups facilitated cell proliferation and migration, inhibited apoptosis in A549/DDP cells, decreased apoptosis, and increased tumor size and growth rate in animal experiments. Immunohistochemistry showed that Ki67 levels increased, and levels of apoptotic indicators significantly decreased in the WDHD1 and MAPRE2 knockout groups. Clinical data confirmed that WDHD1 overexpression negatively correlated with cisplatin sensitivity. Thus, the ubiquitin ligase WDHD1 induces cisplatin resistance in LUAD by promoting MAPRE2 ubiquitination.
Collapse
Affiliation(s)
- Lian Gong
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Mengqing Xiao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Dong He
- Department of Respiratory, The Second People's Hospital of Hunan Province, Changsha, China
| | - Yi Hu
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Yuxing Zhu
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Liang Xiang
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ying Bao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoming Liu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianye Liu
- Department of Urology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ming Zhou
- Cancer Research Institute and Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Central South University, Changsha, China
| | - Yanhong Zhou
- Cancer Research Institute and Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Central South University, Changsha, China
| | - Yaxin Cheng
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Yeyu Zhang
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Liping Deng
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Rongrong Zhu
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Hua Lan
- Department of Gynaecology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
30
|
Sharma A, Trivedi AK. Regulation of apoptosis by E3 ubiquitin ligases in ubiquitin proteasome system. Cell Biol Int 2019; 44:721-734. [PMID: 31814188 DOI: 10.1002/cbin.11277] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/06/2019] [Indexed: 11/10/2022]
Abstract
Apoptosis is an organised ATP-dependent programmed cell death that organisms have evolved to maintain homoeostatic cell numbers and eliminate unnecessary or unhealthy cells from the system. Dysregulation of apoptosis can have serious manifestations culminating into various diseases, especially cancer. Accurate control of apoptosis requires regulation of a wide range of growth enhancing as well as anti-oncogenic factors. Appropriate regulation of magnitude and temporal expression of key proteins is vital to maintain functional apoptotic signalling. Controlled protein turnover is thus critical to the unhindered operation of the apoptotic machinery, disruption of which can have severe consequences, foremost being oncogenic transformation of cells. The ubiquitin proteasome system (UPS) is one such major cellular pathway that maintains homoeostatic protein levels. Recent studies have found interesting links between these two fundamental cellular processes, wherein UPS depending on the cue can either inhibit or promote apoptosis. A diverse range of E3 ligases are involved in regulating the turnover of key proteins of the apoptotic pathway. This review summarises an overview of key E3 ubiquitin ligases involved in the regulation of the fundamental proteins involved in apoptosis, linking UPS to apoptosis and attempts to emphasize the significance of this relationship in context of cancer.
Collapse
Affiliation(s)
- Akshay Sharma
- LSS008, Division of Cancer Biology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Lucknow, 226031, India
| | - Arun K Trivedi
- LSS008, Division of Cancer Biology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Lucknow, 226031, India
| |
Collapse
|
31
|
Li S, Han J, Guo G, Sun Y, Zhang T, Zhao M, Xu Y, Cui Y, Liu Y, Zhang J. Voltage-gated sodium channels β3 subunit promotes tumorigenesis in hepatocellular carcinoma by facilitating p53 degradation. FEBS Lett 2019; 594:497-508. [PMID: 31626714 DOI: 10.1002/1873-3468.13641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 11/08/2022]
Abstract
The voltage-gated sodium channels (VGSCs) are aberrantly expressed in a variety of tumors and play an important role in tumor growth and metastasis. Here, we show that VGSCs auxiliary β3 subunit, encoded by the SCN3B gene, promotes proliferation and suppresses apoptosis in HepG2 cells by promoting p53 degradation. β3 significantly increases HepG2 cell proliferation, promotes tumor growth in mouse xenograft models, and suppresses senescence and apoptosis. We found that β3 knockdown stabilizes p53 protein, leading to potentiation of p53-induced cell cycle arrest, senescence, and apoptosis. Mechanistic studies revealed that β3 could bind to p53, promoting p53 ubiquitination and degradation by stabilizing the p53/MDM2 complex. Our results suggest that β3 is a novel negative regulator of p53 and a potential oncogenic factor.
Collapse
Affiliation(s)
- Shuai Li
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Jiadi Han
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Guili Guo
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Yudi Sun
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Tingting Zhang
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Mingyi Zhao
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Yijia Xu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Yong Cui
- School of Medical Devices, Shenyang Pharmaceutical University, China
| | - Yanfeng Liu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China
| | - Jinghai Zhang
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, China.,School of Medical Devices, Shenyang Pharmaceutical University, China
| |
Collapse
|
32
|
Xue M, Zhang K, Mu K, Xu J, Yang H, Liu Y, Wang B, Wang Z, Li Z, Kong Q, Li X, Wang H, Zhu J, Zhuang T. Regulation of estrogen signaling and breast cancer proliferation by an ubiquitin ligase TRIM56. Oncogenesis 2019; 8:30. [PMID: 31000690 PMCID: PMC6473003 DOI: 10.1038/s41389-019-0139-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 11/13/2022] Open
Abstract
Breast cancer ranks no. 1 in women cancer worldwide, while 60–70% are estrogen receptor alpha positive. The estrogen selective modulators, such as tamoxifen, become the effective drugs for controlling ER alpha breast cancer progression. However, tamoxifen resistance will develop during long-time treatment and cancer progression. Thus, further understanding of ER alpha signaling becomes necessary for the improvement of breast cancer therapy. Here, we identify TRIM56 as a novel regulatory factor in ER alpha signaling. TRIM56 expression is positively correlated with ER alpha and PR in breast cancer samples and is related to poor prognosis in endocrine therapy patients. TRIM56 depletion significantly decreases ER alpha signaling activity and ER-alpha-positive breast cancer proliferation in vitro and in vivo. TRIM56 associates with AF1 domain of ER alpha via its WD40 domain in the cytoplasm. TRIM56 prolongs ER alpha protein stability, possibly through targeting ER alpha K63-linked ubiquitination. In conclusion, our study reveals an interesting posttranslational mechanism between TRIM56 and ER alpha in breast cancer progression. Targeting TRIM56 could be a promising approach for ER-alpha-positive breast cancer.
Collapse
Affiliation(s)
- Min Xue
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Kai Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, 250012, Jinan, Shandong, P.R. China
| | - Kun Mu
- Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, P.R. China
| | - Juntao Xu
- Rhil Rivers Technology (Beijing) Ltd, Beijing, P.R. China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, P.R. China
| | - Huijie Yang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Heping District, 300070, Tianjin, P.R. China
| | - Yun Liu
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Beibei Wang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Zhonghao Wang
- School of Stomatology, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Zhongbo Li
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Qiong Kong
- School of International Education, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Xiumin Li
- Department of Gastroenterology, The Third Affiliated Hospital of Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China.
| | - Jian Zhu
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China. .,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Ting Zhuang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China.
| |
Collapse
|
33
|
Weidle UH, Epp A, Birzele F, Brinkmann U. The Functional Role of Prostate Cancer Metastasis-related Micro-RNAs. Cancer Genomics Proteomics 2019; 16:1-19. [PMID: 30587496 DOI: 10.21873/cgp.20108] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/08/2018] [Accepted: 11/23/2018] [Indexed: 02/06/2023] Open
Abstract
The mortality of patients with hormone-resistant prostate cancer can be ascribed to a large degree to metastasis to distant organs, predominantly to the bones. In this review, we discuss the contribution of micro-RNAs (miRs) to the metastatic process of prostate cancer. The criteria for selection of miRs for this review were the availability of preclinical in vivo metastasis-related data in conjunction with prognostic clinical data. Depending on their function in the metastatic process, the corresponding miRs are up- or down-regulated in prostate cancer tissues when compared to matching normal tissues. Up-regulated miRs preferentially target suppressors of cytokine signaling or tumor suppressor-related genes and metastasis-inhibitory transcription factors. Down-regulated miRs promote epithelial-mesenchymal transition or mesenchymal-epithelial transition and diverse pro-metastatic signaling pathways. Some of the discussed miRs exert their function by simultaneously targeting epigenetic pathways as well as cell-cycle-related, anti-apoptotic and signaling-promoting targets. Finally, we discuss potential therapeutic options for the treatment of prostate cancer-related metastases by substitution or inhibition of miRs.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Alexandra Epp
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
34
|
Yu S, Dai J, Ma M, Xu T, Kong Y, Cui C, Chi Z, Si L, Tang H, Yang L, Sheng X, Guo J. RBCK1 promotes p53 degradation via ubiquitination in renal cell carcinoma. Cell Death Dis 2019; 10:254. [PMID: 30874541 PMCID: PMC6420644 DOI: 10.1038/s41419-019-1488-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 01/05/2023]
Abstract
Renal cell carcinoma (RCC) accounts for approximately 3% of adult malignancies, and the incidence of RCC continues to rise worldwide. Although RCC can be treated with surgery at an early stages, the five-year survival rates have been observed to decline dramatically in patients with advanced disease. Most patients with RCC treated with cytotoxic or targeted drugs will develop resistance at some point during therapy. Thus, it is necessary to identify novel therapeutic targets for RCC. Here, we found that RANBP2-type and C3HC4-type zinc finger-containing 1 (RBCK1) expression was upregulated in human RCC samples. Analysis of multiple public databases revealed the correlation between RBCK1 expression and poor prognosis in RCC patients. Subsequently, we performed RBCK1 depletion experiments in RCC cells that severely affected the in vivo and in vitro proliferation of renal cancer cells. The effects of RBCK1 on cell proliferation could be rescued with p53 expression knockdown in two cell lines expressing wild-type p53. Further experiments demonstrated that RBCK1 could facilitate p53 poly-ubiquitination and degradation by direct interaction with p53. Together, our results show that RBCK1 may serve as a promising target for RCC therapy by restoring p53 functions.
Collapse
Affiliation(s)
- Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Tianxiao Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
35
|
Yu N, Xue M, Wang W, Xia D, Li Y, Zhou X, Pang D, Lu K, Hou J, Zhang A, Zhuang T, Wang L, Chang T, Li X. RNF168 facilitates proliferation and invasion of esophageal carcinoma, possibly via stabilizing STAT1. J Cell Mol Med 2018; 23:1553-1561. [PMID: 30506884 PMCID: PMC6349343 DOI: 10.1111/jcmm.14063] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/28/2022] Open
Abstract
Oesophageal cancer ranks as one of the most common malignancy in China and worldwide. Although genome‐wide association studies and molecular biology studies aim to elucidate the driver molecules in oesophageal cancer progression, the detailed mechanisms remain to be identified. Interestingly, RNF168 (RING finger protein 168) shows a high frequency of gene amplification in oesophageal cancer from TCGA database. Here, we report an important function for RNF168 protein in supporting oesophageal cancer growth and invasion by stabilizing STAT1 protein. RNF168 gene is amplified in oesophageal cancer samples, which tends to correlate with poor prognosis. Depletion RNF168 causes decreased cell proliferation and invasion in oesophageal cancer cells. Through unbiased RNA sequencing in RNF168 depleted oesophageal cancer cell, we identifies JAK‐STAT pathway is dramatically decreased. Depletion RNF168 reduced JAK‐STAT target genes, such as IRF1, IRF9 and IFITM1. Immuno‐precipitation reveals that RNF168 associates with STAT1 in the nucleus, stabilizing STAT1 protein and inhibiting its poly‐ubiquitination and degradation. Our study provides a novel mechanism that RNF168 promoting JAK‐STAT signalling in supporting oesophageal cancer progression. It could be a promising strategy to target RNF168 for oesophageal cancer treatment.
Collapse
Affiliation(s)
- Na Yu
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Min Xue
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Weilong Wang
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Dongxue Xia
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Yajie Li
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Xiaofeng Zhou
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Dan Pang
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Kui Lu
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Jinghan Hou
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Aijia Zhang
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Ting Zhuang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| | - Lidong Wang
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, P.R. China
| | - Tingmin Chang
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, P.R. China
| | - Xiumin Li
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, Henan, P.R. China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan Province, P.R. China
| |
Collapse
|
36
|
Hou Y, Li X, Li Q, Xu J, Yang H, Xue M, Niu G, Zhuo S, Mu K, Wu G, Li X, Wang H, Zhu J, Zhuang T. STAT1 facilitates oestrogen receptor α transcription and stimulates breast cancer cell proliferation. J Cell Mol Med 2018; 22:6077-6086. [PMID: 30334368 PMCID: PMC6237559 DOI: 10.1111/jcmm.13882] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022] Open
Abstract
Oestrogen receptor α (ERα) is overexpressed in two‐thirds of all breast cancer cases and is involved in breast cancer development and progression. Although ERα ‐positive breast cancer can be effectively treated by endocrine therapy, endocrine resistance is an urgent clinical problem. Thus, further understanding of the underlying mechanisms involved in ERα signalling is critical in dealing with endocrine resistance in patients with breast cancer. In the present study, unbiased RNA sequence analysis was conducted between the MCF‐7 and MCF‐7 tamoxifen‐resistant (LCC2) cell lines in order to identify differentially expressed genes. The whole transcriptomic data indicated that the JAK‐STAT pathway is markedly up‐regulated, particularly the ISGF3 complex. As the critical effectors, STAT1 and IRF9 were up‐regulated 5‐ and 20‐fold, respectively, in LCC2 cells. The biological experiments indicated that STAT1 is important for ERα signalling. Depletion of STAT1 or inhibition of STAT1 function significantly decreased levels of ERα protein, ERα ‐target gene expression and cell proliferation in both the MCF‐7 and LCC2 cell lines. Chromatin immunoprecipitation revealed that ERα transcription is associated with STAT1 recruitment to the ERα promoter region, suggesting that transcriptional regulation is one mechanism by which STAT1 regulates ERα mRNA levels and ERα signalling in breast cancer cells. The present study reveals a possible endocrine‐resistant mechanism by which STAT1 modulates ERα signalling and confers tamoxifen resistance. Targeting of STAT1 is a potential treatment strategy for endocrine‐resistant breast cancers.
Collapse
Affiliation(s)
- Yingxiang Hou
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan, China
| | - Xin Li
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan, China
| | - Qianhua Li
- Department of Pathology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Juntao Xu
- Rhil Rivers Technology (Beijing) Ltd, Beijing, China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, China
| | - Huijie Yang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan, China
| | - Min Xue
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan, China
| | - Gang Niu
- Rhil Rivers Technology (Beijing) Ltd, Beijing, China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, China
| | - Shu Zhuo
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Kun Mu
- Department of Pathology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Xiumin Li
- Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hui Wang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jian Zhu
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Ting Zhuang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
37
|
Sun S, Wu Q, Song J, Sun S. Protein kinase C δ-dependent regulation of Ubiquitin-proteasome system function in breast cancer. Cancer Biomark 2018; 21:1-9. [PMID: 29036789 DOI: 10.3233/cbm-170451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Besides the crucial role of hyperinsulinemia in the development of breast cancer with Type 2 diabetes mellitus (T2DM), it has been shown that hyperglycemia could contribute to promote cancer progression. A remarkable association within hyperglycemia, PKCδ and Ubiquitin-proteasome system (UPS) has been reported, suggesting that PKCδ may mediate high glucose-induced UPS activation in breast cancer cells. Although the independent effects of PKCδ or UPS on breast cancer and T2DM are increasingly supported by experimental evidence, the complex interactional link between PKCδ and UPS is still unclear. Hence, we focus on the relationship between PKCδ and UPS in breast cancer with T2DM. We hypothesize that PKCδ may have the function to regulate the activity of UPS. Further, we speculate that PKCδ combine with proteasome α2 promoter, that indicate PKCδ regulate the function of UPS by change the composition of proteasome. Therefore, we surmise that PKCδ mediated high glucose-induced UPS activation in breast cancer cells, and specific PKCδ inhibitor rottlerin significantly suppressed elevated glucose induced the activity of UPS. We hope that our paper will stimulate further studies the relationship between PKCδ and UPS, and a new targeted therapy and early medical intervention for PKCδ could be a useful option for breast cancer cases complicated with T2DM or hyperglycemia.
Collapse
Affiliation(s)
- Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junlong Song
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
38
|
Liu Z, Zhang J, Xu J, Yang H, Li X, Hou Y, Zhao Y, Xue M, Wang B, Yu N, Yu S, Niu G, Wu G, Li X, Wang H, Zhu J, Zhuang T. RNF168 facilitates oestrogen receptor ɑ transcription and drives breast cancer proliferation. J Cell Mol Med 2018; 22:4161-4170. [PMID: 29974997 PMCID: PMC6111850 DOI: 10.1111/jcmm.13694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/23/2018] [Indexed: 01/13/2023] Open
Abstract
Oestrogen receptor ɑ (ERɑ) is overexpressed in two-thirds of all breast cancers and involves in development and breast cancer progression. Although ERɑ-positive breast cancer could be effective treated by endocrine therapy, the endocrine resistance is still an urgent clinical problem. Thus, further understanding of the underlying mechanisms ERɑ signalling is critical in dealing with endocrine resistance in breast cancer patients. MCF-7 and T47D breast cancer cell lines are used to carry out the molecular biological experiments. Western blot is used to assess the relative protein level of ERɑ, RNF168 and actin. Real-time PCR is used the measure the relative ERɑ-related gene mRNA level. Luciferase assay is used to measure the relative ERɑ signalling activity. Chromatin immunoprecipitation is used to measure the RNF168 binding affinity to ERɑ promoter regions. WST assay and flow cytometry are used to measure the cell proliferation capacity. We use Student's t test and one-way ANOVA test for statistical data analysis. Here, we report an important role in ERɑ-positive breast cancer cells for RNF168 protein in supporting cell proliferation by driving the transcription of ERɑ. RNF168 is highly expressed in breast cancer samples, compared with normal breast tissue. In patients with breast cancer, RNF168 expression level is correlated with poor endocrine treatment outcome. Depletion of RNF168 causes decreased cell proliferation in MCF-7 and T47D cells. Besides, depletion RNF168 reduced mRNA level of ERɑ and its target genes, such as PS2 and GREB1. Chromatin immunoprecipitation revealed that ERɑ transcription is associated with RNF168 recruitment to ERɑ promoter region, suggesting that transcriptional regulation is one mechanism by which RNF168 regulates ERɑ mRNA level and ERɑ signalling in breast cancer cells. RNF168 is required for ERɑ-positive breast cancer cell proliferation and facilitate ERɑ signalling activity possibly through promoting transcription of ERɑ.
Collapse
Affiliation(s)
- Zhenhua Liu
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China.,Synthetic Biology Engineering Lab of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jinghang Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Juntao Xu
- Rhil Rivers Technology (Beijing) Ltd., Beijing, China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, China
| | - Huijie Yang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Xin Li
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Yingxiang Hou
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Yan Zhao
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Min Xue
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Beibei Wang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Na Yu
- Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, China
| | - Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal cancer and Melanoma, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Gang Niu
- Rhil Rivers Technology (Beijing) Ltd., Beijing, China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiumin Li
- Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, China
| | - Hui Wang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China
| | - Jian Zhu
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ting Zhuang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
39
|
Yang H, Yu N, Xu J, Ding X, Deng W, Wu G, Li X, Hou Y, Liu Z, Zhao Y, Xue M, Yu S, Wang B, Li X, Niu G, Wang H, Zhu J, Zhuang T. SMURF1 facilitates estrogen receptor ɑ signaling in breast cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:24. [PMID: 29433542 PMCID: PMC5808446 DOI: 10.1186/s13046-018-0672-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/02/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Estrogen receptor alpha (ER alpha) is expressed in the majority of breast cancers and promotes estrogen-dependent cancer progression. ER alpha positive breast cancer can be well controlled by ER alpha modulators, such as tamoxifen. However, tamoxifen resistance is commonly observed by altered ER alpha signaling. Thus, further understanding of the molecular mechanisms, which regulates ER alpha signaling, is important to improve breast cancer therapy. METHODS SMURF1 and ER alpha protein expression levels were measured by western blot, while the ER alpha target genes were measured by real-time PCR. WST-1 assay was used to measure cell viability; the xeno-graft tumor model were used for in vivo study. RNA sequencing was analyzed by Ingenuity Pathway Analysis. Identification of ER alpha signaling was accomplished with luciferase assays, real-time RT-PCR and Western blotting. Protein stability assay and ubiquitin assay was used to detect ER alpha protein degradation. Immuno-precipitation based assays were used to detect the interaction domain between ER alpha and SMURF1. The ubiquitin-based Immuno-precipitation based assays were used to detect the specific ubiquitination manner happened on ER alpha. RESULTS Here, we identify the E3 ligase SMURF1 facilitates ER alpha signaling. We show that depletion SMURF1 decreases ER alpha positive cell proliferation in vitro and in vivo. SMURF1 depletion based RNA-sequence data shows SMURF1 is necessary for ER alpha target gene expression in the transcriptomic scale. Immunoprecipitation indicates that SMURF1 associates with the N-terminal of ER alpha in the cytoplasm via its HECT domain. SMURF1 increases ER alpha stability, possibly by inhibiting K48-specific poly-ubiquitination process on ER alpha protein. Interestingly, SMURF1 expression could be induced via estradiol treatment. CONCLUSIONS Our study reveals a novel positive feedback between SMURF1 and ER alpha signaling in supporting breast cancer growth. Targeting SMURF1 could be one promising strategy for ER alpha positive breast cancer treatment.
Collapse
Affiliation(s)
- Huijie Yang
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Na Yu
- Department of Gastroenterology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Juntao Xu
- Phil Rivers Technology (Beijing) Ltd, Beijing, China.,Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd, Shenzhen, China
| | - Xiaosheng Ding
- Department of Medical Oncology, Peking University International Hospital, Beijing, China
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center of Digestive Diseases, Beijing, 100050, China
| | - Guojin Wu
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xin Li
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Yingxiang Hou
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Zhenhua Liu
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Yan Zhao
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Min Xue
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Department of Renal cancer and Melanoma, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Beibei Wang
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Xiumin Li
- Department of Gastroenterology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, China
| | - Gang Niu
- Phil Rivers Technology (Beijing) Ltd, Beijing, China. .,Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd, Shenzhen, China.
| | - Hui Wang
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China. .,Xinxiang Medical University, School of Laboratory Medicine, Xinxiang, Henan Province, China.
| | - Jian Zhu
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China. .,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Ting Zhuang
- Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China. .,Xinxiang Medical University, School of Laboratory Medicine, Xinxiang, Henan Province, China.
| |
Collapse
|
40
|
Clinorotation-induced autophagy via HDM2-p53-mTOR pathway enhances cell migration in vascular endothelial cells. Cell Death Dis 2018; 9:147. [PMID: 29396411 PMCID: PMC5833810 DOI: 10.1038/s41419-017-0185-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/14/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Abstract
Individuals exposed to long-term spaceflight often experience cardiovascular dysfunctions characterized by orthostatic intolerance, disability on physical exercise, and even frank syncope. Recent studies have showed that the alterations of cardiovascular system are closely related to the functional changes of endothelial cells. We have shown previously that autophagy can be induced by simulated microgravity in human umbilical vein endothelial cells (HUVECs). However, the mechanism of enhanced autophagy induced by simulated microgravity and its role in the regulation of endothelial function still remain unclear. We report here that 48 h clinorotation promoted cell migration in HUVECs by induction of autophagy. Furthermore, clinorotation enhanced autophagy by the mechanism of human murine double minute 2 (HDM2)-dependent degradation of cytoplasmic p53 at 26S proteasome, which results in the suppression of mechanistic target of rapamycin (mTOR), but not via activation of AMPK in HUVECs. These results support the key role of HDM2–p53 in direct downregulation of mTOR, but not through AMPK in microgravity-induced autophagy in HUVECs.
Collapse
|
41
|
Wang L, Hou Z, Hasim A, Abuduerheman A, Zhang H, Niyaz M, Awut I, Upur H, Sheyhidin I. RNF113A promotes the proliferation, migration and invasion, and is associated with a poor prognosis of esophageal squamous cell carcinoma. Int J Oncol 2018; 52:861-871. [PMID: 29393393 DOI: 10.3892/ijo.2018.4253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 12/04/2017] [Indexed: 11/06/2022] Open
Abstract
Ring finger protein 113A (RNF113A) possesses a C3HC4 zinc finger domain and this domain is found in E3 ubiquitin ligase and is involved in tumorigenesis. To date, and at least to the best of our knowledge, there are no studies available which have investigated RNF113A in cancer. Thus, this study aimed to explore the role of RNF113A in the development of esophageal squamous cell carcinoma (ESCC). For this purpose, paraffin-embedded samples from 117 patients with ESCC were selected, as well as 41 pairs of fresh-frozen ESCC and adjacent normal tissue samples. RNF113A expression was examined by immunohistochemistry and reverse transcription-quantitative PCR (RT-qPCR). RNF113A was overexpressed or silenced in the EC9706 and Eca109 cells. The cells were examined for cell cycle progression, apoptosis, invasiveness and migration. Xenograft tumors were also created in mice using the Eca109 cells. Tumor differentiation (P=0.008) and T classification (P<0.001) were found to be significantly associated with RNF113A expression. No statistically significant association was observed between RNF113A expression and sex, age, histological type, tumor location and lymph node metastasis (N classification). Kaplan-Meier analysis revealed that the patients with ESCC with ahigh expression of RNF113A had a lower survival rate than those with a low expression (P=0.002). Multivariate analysis revealed that RNF113A expression (HR=2.406; 95% CI, 1.301-4.449, P=0.005) was independently associated with overall survival in patients with ESCC. The overexpression of RNF113A promoted proliferation, migration, and invasiveness of ESCC cell lines in vitro, and RNF113A silencing reversed these malignant behaviors. RNF113A knockdown inhibited tumor growth in vivo. Thus, these results indicate that RNF113A promotes the proliferation, migration and invasiveness of ESCC cell lines. RNF113A expression in ESCC is this associated with a poor prognosis of affected patients.
Collapse
Affiliation(s)
- Lei Wang
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Zhichao Hou
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Ayshamgul Hasim
- Department of Pathology, Medical University of Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Abulajiang Abuduerheman
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Haiping Zhang
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Madiniyat Niyaz
- Clinical Medical Research Institute, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Idiris Awut
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Halmurat Upur
- Department of Uyghur Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| | - Ilyar Sheyhidin
- Department of Thoracic Surgery, Τhe First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830054, P.R. China
| |
Collapse
|
42
|
Qiu P, Xu TJ, Lu XD, Yang W, Zhang YB, Xu GM. MicroRNA-378 regulates cell proliferation and migration by repressing RNF31 in pituitary adenoma. Oncol Lett 2018; 15:789-794. [PMID: 29399147 PMCID: PMC5772874 DOI: 10.3892/ol.2017.7431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/07/2017] [Indexed: 12/25/2022] Open
Abstract
MicroRNA-378 (miR-378) is dysregulated in multiple malignancies and is associated with tumor progression. However, the expression and mechanism of miR-378 in pituitary adenoma (PA) remains to be elucidated. In the present study, the role and mechanism of miR-378 in PA tumorigenesis and development was investigated. It was revealed that the levels of miR-378 expression were markedly downregulated in PA tissues. CCK-8 and wound healing assays revealed that transfection with miR-378 mimics was able to markedly inhibit the proliferation and migration of GH3 cells. Furthermore, quantitative polymerase chain reaction analysis demonstrated that ring finger protein 31 (RNF31) was upregulated in PA specimens and the levels of RNF31 expression was negatively regulated by miR-378. In addition, knockdown of RNF31 markedly suppressed cell proliferation and migration in GH3 cells. In conclusion, the present study provides a molecular basis for the function of miR-378/RNF31 in the progression of human PA, indicating a potential novel target for the treatment of PA.
Collapse
Affiliation(s)
- Peng Qiu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Tong-Jiang Xu
- Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, Shandong 271199, P.R. China
| | - Xiang-Dong Lu
- Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, Shandong 271199, P.R. China
| | - Wei Yang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yu-Bao Zhang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Guang-Ming Xu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
43
|
Functional significance and therapeutic implication of ring-type E3 ligases in colorectal cancer. Oncogene 2017; 37:148-159. [PMID: 28925398 PMCID: PMC5770599 DOI: 10.1038/onc.2017.313] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/29/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023]
Abstract
Accumulative studies revealed that E3 ubiquitin ligases have important roles in colorectal carcinogenesis. The pathogenic mechanisms of colorectal cancer (CRC) initiation and progression are complex and heterogeneous, involving somatic mutations, abnormal gene fusion, deletion or amplification and epigenetic alteration, which may cause aberrant expression or altered function of E3 ligases in CRC. Defects of E3 ligases have been reported to be involved in the molecular etiology and pathogenesis of CRC. The aberrant expressed E3 ligases can function as either oncogenes or tumor suppressors depending on ubiquiting target substrates in CRC. Recently, considerable progress has been made in our understanding of the potential roles of E3 ligase-mediated ubiquitylation in colorectal carcinogenesis. There are mainly two subtypes of E3 ubiquitin ligases in humans, as defined by the presence of either a HECT domain or a RING finger domain on the basis of structural similitude. Most cancer-associated E3 ligases participate in regulating the cell cycle, apoptosis, gene transcription, cell signaling and DNA repair, the critical parts of CRC tumorigenesis. In this review, we have provided a comprehensive summary of abnormally expressed E3 ligases and their related pivotal mechanistic effects in CRC. In particular, we have highlighted the function of RING-type E3 ubiquitin enzymes in modulating cancer signaling pathways, immunity and tumor microenvironment in CRC development and progression; their mechanism(s) of action in CRC involving both ubiquitylation-dependent and ubiquitylation-independent effects; and the potential of RING E3 ligases as molecular biomarkers for predicting patient prognosis and as therapeutic targets in CRC. A better understanding of E3 ligase-mediated substrates' ubiquitylation involved in the development of CRC will provide new insights into the pathophysiology mechanisms of CRC, and unravel novel prognostic markers and therapeutic strategies for CRC.
Collapse
|
44
|
Zhuang T, Yu S, Zhang L, Yang H, Li X, Hou Y, Liu Z, Shi Y, Wang W, Yu N, Li A, Li X, Li X, Niu G, Xu J, Hasni MS, Mu K, Wang H, Zhu J. SHARPIN stabilizes estrogen receptor α and promotes breast cancer cell proliferation. Oncotarget 2017; 8:77137-77151. [PMID: 29100376 PMCID: PMC5652769 DOI: 10.18632/oncotarget.20368] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/29/2017] [Indexed: 12/26/2022] Open
Abstract
Estrogen receptor α is expressed in the majority of breast cancers and promotes estrogen-dependent cancer progression. In our study, we identified the novel E3 ubiquitin ligase SHARPIN function to facilitate ERα signaling. SHARPIN is highly expressed in human breast cancer and correlates with ERα protein level by immunohistochemistry. SHARPIN expression level correlates with poor prognosis in ERα positive breast cancer patients. SHARPIN depletion based RNA-sequence data shows that ERα signaling is a potential SHARPIN target. SHARPIN depletion significantly decreases ERα protein level, ERα target genes expression and estrogen response element activity in breast cancer cells, while SHARPIN overexpression could reverse these effects. SHARPIN depletion significantly decreases estrogen stimulated cell proliferation in breast cancer cells, which effect could be further rescued by ERα overexpression. Further mechanistic study reveals that SHARPIN mainly localizes in the cytosol and interacts with ERα both in the cytosol and the nuclear. SHARPIN regulates ERα signaling through protein stability, not through gene expression. SHARPIN stabilizes ERα protein via prohibiting ERα protein poly-ubiquitination. Further study shows that SHARPIN could facilitate the mono-ubiquitinaiton of ERα at K302/303 sites and facilitate ERE luciferase activity. Together, our findings propose a novel ERα modulation mechanism in supporting breast cancer cell growth, in which SHARPIN could be one suitable target for development of novel therapy for ERα positive breast cancer.
Collapse
Affiliation(s)
- Ting Zhuang
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Sifan Yu
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Renal Cancer and Melanoma, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Lichen Zhang
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Laboratory of Genetic Regulators in the Immune System, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Huijie Yang
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xin Li
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yingxiang Hou
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhenhua Liu
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Synthetic Biology Remaking Engineering and Application Laboratory, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yuanyuan Shi
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Weilong Wang
- Department of Gastroenterology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, China
| | - Na Yu
- Department of Gastroenterology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, China
| | - Anqi Li
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,School of International Education, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xuefeng Li
- Department of Medical Oncology, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Xiumin Li
- Department of Gastroenterology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, China
| | - Gang Niu
- Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd, Shenzhen, Guangdong, China.,Phil Rivers Technology (Beijing) Ltd. Beijing, China.,Institute of Biochemistry University of Balochistan, Quetta, Pakistan
| | - Juntao Xu
- Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd, Shenzhen, Guangdong, China.,Phil Rivers Technology (Beijing) Ltd. Beijing, China.,Institute of Biochemistry University of Balochistan, Quetta, Pakistan
| | - Muhammad Sharif Hasni
- Institute of Biochemistry University of Balochistan, Quetta, Pakistan.,Department of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Kun Mu
- Department of Pathology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Wang
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jian Zhu
- Research Center for Immunology, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
45
|
Geng R, Tan X, Zuo Z, Wu J, Pan Z, Shi W, Liu R, Yao C, Wang G, Lin J, Qiu L, Huang W, Chen S. Synthetic lethal short hairpin RNA screening reveals that ring finger protein 183 confers resistance to trametinib in colorectal cancer cells. CHINESE JOURNAL OF CANCER 2017; 36:63. [PMID: 28756770 PMCID: PMC5535279 DOI: 10.1186/s40880-017-0228-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/16/2017] [Indexed: 01/06/2023]
Abstract
Background The mitogen-activated extracellular signal-regulated kinase 1/2 (MEK1/2) inhibitor trametinib has shown promising therapeutic effects on melanoma, but its efficacy on colorectal cancer (CRC) is limited. Synthetic lethality arises with a combination of two or more separate gene mutations that causes cell death, whereas individual mutations keep cells alive. This study aimed to identify the genes responsible for resistance to trametinib in CRC cells, using a synthetic lethal short hairpin RNA (shRNA) screening approach. Methods We infected HT29 cells with a pooled lentiviral shRNA library and applied next-generation sequencing to identify shRNAs with reduced abundance after 8-day treatment of 20 nmol/L trametinib. HCT116 and HT29 cells were used in validation studies. Stable ring finger protein 183 (RNF183)-overexpressing cell lines were generated by pcDNA4-myc/his-RNF183 transfection. Stable RNF183-knockdown cell lines were generated by infection of lentiviruses that express RNF183 shRNA, and small interference RNA (siRNA) was used to knock down RNF183 transiently. Quantitative real-time PCR was used to determine the mRNA expression. Western blotting, immunohistochemical analysis, and enzyme-linked immunosorbent assay (ELISA) were used to evaluate the protein abundance. MTT assay, colony formation assay, and subcutaneous xenograft tumor growth model were used to evaluate cell proliferation. Results In the primary screening, we found that the abundance of RNF183 shRNA was markedly reduced after treatment with trametinib. Trametinib induced the expression of RNF183, which conferred resistance to drug-induced cell growth repression and apoptotic and non-apoptotic cell deaths. Moreover, interleukin-8 (IL-8) was a downstream gene of RNF183 and was required for the function of RNF183 in facilitating cell growth. Additionally, elevated RNF183 expression partly reduced the inhibitory effect of trametinib on IL-8 expression. Finally, xenograft tumor model showed the synergism of RNF183 knockdown and trametinib in repressing the growth of CRC cells in vivo. Conclusion The RNF183-IL-8 axis is responsible for the resistance of CRC cells to the MEK1/2 inhibitor trametinib and may serve as a candidate target for combined therapy for CRC. Electronic supplementary material The online version of this article (doi:10.1186/s40880-017-0228-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rong Geng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Xin Tan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Zhixiang Zuo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Jiangxue Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Zhizhong Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Wei Shi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Ranyi Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Chen Yao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Gaoyuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Jiaxin Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Lin Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China.,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Wenlin Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China. .,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China. .,Guangdong Provincial Key Laboratory of Tumor-Targeted Drugs and Guangzhou Enterprise Key Laboratory of Gene Medicine, Guangzhou Doublle Bioproducts Co. Ltd., Guangzhou, 510507, Guangdong, P. R. China.
| | - Shuai Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510507, Guangdong, P. R. China. .,Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China. .,Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
| |
Collapse
|
46
|
Liu Y, Han L, Bai Y, Du W, Yang B. Down-regulation of MicroRNA-133 predicts poor overall survival and regulates the growth and invasive abilities in glioma. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:206-210. [PMID: 28376685 DOI: 10.1080/21691401.2017.1304551] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
miRNAs were reported as oncogene or tumour suppressors in various cancers and played important roles in tumour development and progression. Dysregulated miR-133 has been reported in several cancers, however, the expression and biological function of miR-133 in glioma remained unclear. In this study, we found that miR-133 expression level was significantly decreased in glioma tissues and cell lines by RT-qPCR. Then miR-133 mimics were used to evaluate the effects of miR-133 on cell proliferation and invasion in vitro. We found that overexpressed miR-133 could significantly suppress cell growth, and invasion in U87 cells. Additionally, we found that forkhead box C1 (FOXC1) was overexpressed in glioma tissue and it was directly regulated by miR-133. Overall, this study is the first proof to demonstrate that miR-133 function as tumour suppressor in glioma and inhibit cell proliferation and invasioned by directly targeting FOXC1, implying miR-133 as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Yu Liu
- a The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Lili Han
- b Cancer Hospital of Henan Province, The Affiliated Cancer Hospital of Zhengzhou University , Zhengzhou , China.,c The Affiliated Cancer Hospital of Zhengzhou University , Zhengzhou , China
| | - Yahui Bai
- a The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Wei Du
- a The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Bo Yang
- a The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
47
|
Yang H, Yu S, Wang W, Li X, Hou Y, Liu Z, Shi Y, Mu K, Niu G, Xu J, Wang H, Zhu J, Zhuang T. SHARPIN Facilitates p53 Degradation in Breast Cancer Cells. Neoplasia 2017; 19:84-92. [PMID: 28063307 PMCID: PMC5219588 DOI: 10.1016/j.neo.2016.12.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/01/2016] [Accepted: 12/05/2016] [Indexed: 11/30/2022]
Abstract
The ubiquitin binding protein SHAPRIN is highly expressed in human breast cancer, one of the most frequent female malignancies worldwide. Here, we perform SHARPIN depletion in breast cancer cells together with RNA sequencing. The global expression profiling showed p53 signaling as a potential SHARPIN target. SHARPIN depletion decreased cell proliferation, which effect could be rescue by p53 knocking down. Depletion SHARPIN significantly increases p53 protein level and its target genes in multiple breast cancer cell lines. Further experiment revealed that SHARPIN could facilitate p53 poly-ubiquitination and degradation in MDM2 dependent manner. Immuno-precipitation assay showed that SHARPIN associated with MDM2 and prolonged MDM2 protein stability. Analysis of public available database showed SHARPIN correlated with poor prognosis specifically in p53 wild-type breast cancer patients. Together, our finding revealed a novel modifier for p53/MDM2 complex and suggested SHARPIN as a promising target to restore p53 function in breast cancer.
Collapse
Affiliation(s)
- Huijie Yang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Sifan Yu
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Department of Renal Cancer and Melanoma, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Weilong Wang
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Xin Li
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Yingxiang Hou
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Zhenhua Liu
- College of Life Science and Technology, Synthetic Biology, Medical Institute, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Yuanyuan Shi
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Kun Mu
- Department of Pathology, Shandong University School of Medicine, Jinan, PR China
| | - Gang Niu
- Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd., Shenzhen, PR China
| | - Juntao Xu
- Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd., Shenzhen, PR China
| | - Hui Wang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China.
| | - Jian Zhu
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China.
| | - Ting Zhuang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China.
| |
Collapse
|
48
|
Zhuang T, Zhu J, Li Z, Lorent J, Zhao C, Dahlman-Wright K, Strömblad S. p21-activated kinase group II small compound inhibitor GNE-2861 perturbs estrogen receptor alpha signaling and restores tamoxifen-sensitivity in breast cancer cells. Oncotarget 2016; 6:43853-68. [PMID: 26554417 PMCID: PMC4791272 DOI: 10.18632/oncotarget.6081] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/26/2015] [Indexed: 01/07/2023] Open
Abstract
Estrogen receptor alpha (ERα) is highly expressed in most breast cancers. Consequently, ERα modulators, such as tamoxifen, are successful in breast cancer treatment, although tamoxifen resistance is commonly observed. While tamoxifen resistance may be caused by altered ERα signaling, the molecular mechanisms regulating ERα signaling and tamoxifen resistance are not entirely clear. Here, we found that PAK4 expression was consistently correlated to poor patient outcome in endocrine treated and tamoxifen-only treated breast cancer patients. Importantly, while PAK4 overexpression promoted tamoxifen resistance in MCF-7 human breast cancer cells, pharmacological treatment with a group II PAK (PAK4, 5, 6) inhibitor, GNE-2861, sensitized tamoxifen resistant MCF-7/LCC2 breast cancer cells to tamoxifen. Mechanistically, we identified a regulatory positive feedback loop, where ERα bound to the PAK4 gene, thereby promoting PAK4 expression, while PAK4 in turn stabilized the ERα protein, activated ERα transcriptional activity and ERα target gene expression. Further, PAK4 phosphorylated ERα-Ser305, a phosphorylation event needed for the PAK4 activation of ERα-dependent transcription. In conclusion, PAK4 may be a suitable target for perturbing ERα signaling and tamoxifen resistance in breast cancer patients.
Collapse
Affiliation(s)
- Ting Zhuang
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Jian Zhu
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Zhilun Li
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Julie Lorent
- Department of Oncology and Pathology, Karolinska Institutet, Solna, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden.,Science for Life Laboratory (SciLifeLab), Karolinska Institutet, Solna, Sweden
| | - Staffan Strömblad
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
49
|
Zhu J, Zhuang T, Yang H, Li X, Liu H, Wang H. Atypical ubiquitin ligase RNF31: the nuclear factor modulator in breast cancer progression. BMC Cancer 2016; 16:538. [PMID: 27460922 PMCID: PMC4962416 DOI: 10.1186/s12885-016-2575-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 07/18/2016] [Indexed: 12/16/2022] Open
Abstract
Breast cancer causes the No.1 women cancer prevalence and the No.2 women cancer mortality worldwide. Nuclear receptor/transcriptional factor signaling is aberrant and plays important roles in breast cancer pathogenesis and evolution, such as estrogen receptor α (ERα/ESR1), tumor protein p53 (p53/TP53) and Nuclear factor kappa B (NFκB). About 60–70 % of breast tumors are ERα positive, while approximate 70 % of breast tumors are P53 wild type. Recent studies indicate that nuclear receptors/transcriptional factors could be tightly controlled through protein post-translational modification. The nuclear receptors/transcriptional factors could endure several types of modifications, including phosphorylation, acetylation and ubiquitination. Compared with the other two types of modifications, ubiquitination was mostly linked to protein degradation process, while few researches focused on the functional changes of the target proteins. Until recent years, ubiquitination process is no longer regarded as merely a protein degradation process, but aslo treated as one kind of modification signal. As an atypical E3 ubiquitin ligase, RNF31 was previously found to facilitate NFκB signaling transduction through linear ubiquitination on IKKγ(IκB kinase γ). Our previous studies showed important regulatory functions of RNF31 in controlling important oncogenic pathways in breast cancer, such as ERα and p53. This review highlights recent discoveries on RNF31 functions in nuclear factor modifications, breast cancer progression and possible therapeutic inhibitors targeting RNF31.
Collapse
Affiliation(s)
- Jian Zhu
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China. .,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Ting Zhuang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Huijie Yang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Xin Li
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Huandi Liu
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Hui Wang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China.
| |
Collapse
|