1
|
Liu K, Hao X, Gao Y, Cao Z, Hou M, Qin L, Song Y, Wang M, Jiang B, Liu Q, Zou Y, Gong Y, Liu G, Sun G. CUL4B protects kidneys from acute injury by restraining p53/PAI-1 signaling. Cell Death Dis 2024; 15:915. [PMID: 39695153 DOI: 10.1038/s41419-024-07299-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Acute kidney injury (AKI) caused by nephrotoxins, ischemia reperfusion (IR) or sepsis is associated with high morbidity and mortality. Unveiling new mechanisms underlying AKI can help develop new therapeutic strategy. Cullin 4B (CUL4B) is a scaffold protein in the CUL4B-RING E3 ubiquitin ligase (CRL4B) complex. Here, we demonstrate that CUL4B can protect kidneys from acute injury induced by cisplatin and IR. CUL4B is upregulated in mouse tubular epithelial cells (TECs) after cisplatin treatment or IR. Loss of CUL4B in kidneys exacerbates renal injury, inflammation, and apoptosis of TECs caused by cisplatin and IR. Transcriptome analysis reveals that Cul4b deficiency enhances injury-induced PAI-1 expression. CUL4B suppresses PAI-1 expression by promoting polyubiquitination and degradation of p53. Inhibition of either PAI-1 or p53 can prevent the aggravated renal injury and inflammation caused by loss of CUL4B. Our work has identified the kidney-protective role of CUL4B against acute injury.
Collapse
Affiliation(s)
- Kaixuan Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaoyu Hao
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yangfan Gao
- Department of Nephrology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhiyuan Cao
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Min Hou
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lining Qin
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yu Song
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Molin Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Baichun Jiang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qiao Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yongxin Zou
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Guangyi Liu
- Department of Nephrology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Gongping Sun
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Hu Y, Yan Y, Wang J, Hou J, Lin Q. Molecular glue degrader for tumor treatment. Front Oncol 2024; 14:1512666. [PMID: 39759140 PMCID: PMC11697593 DOI: 10.3389/fonc.2024.1512666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/19/2024] [Indexed: 01/07/2025] Open
Abstract
Targeted Protein Degradation (TPD) represented by Proteolysis-Targeting Chimeras (PROTAC) is the frontier field in the research and development of antitumor therapy, in which oral drug HP518 Receives FDA Proceed Authorization for its IND Application for Prostate Cancer Treatment. Recently, molecular glue, functioning via degradation of the target protein is emerging as a promising modality for the development of therapeutic agents, while exhibits greater advantages over PROTAC, including improved efficiency, resistance-free properties, and the capacity to selectively target "undruggable" proteins. This marks a revolutionary advancement in the landscape of small molecule drugs. Given that molecular glue research is still in its early stage, we summarized the mechanisms of molecular glue, the promising drugs in clinical trials and diverse feasible design strategies for molecular glue therapeutics.
Collapse
Affiliation(s)
- Yuhan Hu
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yan Yan
- Department of Infectious Diseases, Zhoukou Central Hospital, Zhoukou, China
| | - Jiehao Wang
- Department of Gastroenterology, Zhengzhou First People's Hospital, Zhengzhou, China
| | - Jiangxue Hou
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Quande Lin
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
3
|
Borlak J, Ciribilli Y, Bisio A, Selvaraj S, Inga A, Oh JH, Spanel R. The Abl1 tyrosine kinase is a key player in doxorubicin-induced cardiomyopathy and its p53/p73 cell death mediated signaling differs in atrial and ventricular cardiomyocytes. J Transl Med 2024; 22:845. [PMID: 39285385 PMCID: PMC11403941 DOI: 10.1186/s12967-024-05623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/16/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Doxorubicin is an important anticancer drug, however, elicits dose-dependently cardiomyopathy. Given its mode of action, i.e. topoisomerase inhibition and DNA damage, we investigated genetic events associated with cardiomyopathy and searched for mechanism-based possibilities to alleviate cardiotoxicity. We treated rats at clinically relevant doses of doxorubicin. Histopathology and transmission electron microscopy (TEM) defined cardiac lesions, and transcriptomics unveiled cardiomyopathy-associated gene regulations. Genomic-footprints revealed critical components of Abl1-p53-signaling, and EMSA-assays evidenced Abl1 DNA-binding activity. Gene reporter assays confirmed Abl1 activity on p53-targets while immunohistochemistry/immunofluorescence microscopy demonstrated Abl1, p53&p73 signaling. RESULTS Doxorubicin treatment caused dose-dependently toxic cardiomyopathy, and TEM evidenced damaged mitochondria and myofibrillar disarray. Surviving cardiomyocytes repressed Parkin-1 and Bnip3-mediated mitophagy, stimulated dynamin-1-like dependent mitochondrial fission and induced anti-apoptotic Bag1 signaling. Thus, we observed induced mitochondrial biogenesis. Transcriptomics discovered heterogeneity in cellular responses with minimal overlap between treatments, and the data are highly suggestive for distinct cardiomyocyte (sub)populations which differed in their resilience and reparative capacity. Genome-wide footprints revealed Abl1 and p53 enriched binding sites in doxorubicin-regulated genes, and we confirmed Abl1 DNA-binding activity in EMSA-assays. Extraordinarily, Abl1 signaling differed in the heart with highly significant regulations of Abl1, p53 and p73 in atrial cardiomyocytes. Conversely, in ventricular cardiomyocytes, Abl1 solely-modulated p53-signaling that was BAX transcription-independent. Gene reporter assays established Abl1 cofactor activity for the p53-reporter PG13-luc, and ectopic Abl1 expression stimulated p53-mediated apoptosis. CONCLUSIONS The tyrosine kinase Abl1 is of critical importance in doxorubicin induced cardiomyopathy, and we propose its inhibition as means to diminish risk of cardiotoxicity.
Collapse
Affiliation(s)
- Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Yari Ciribilli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessandra Bisio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Saravanakumar Selvaraj
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Alberto Inga
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
4
|
Liu S, Zhuo L, Chen L, He Y, Chen X, Zhang H, Zhou Y, Ni Z, Zhao S, Hu X. E3 ubiquitin ligase RNF148 functions as an oncogene in colorectal cancer by ubiquitination-mediated degradation of CHAC2. Carcinogenesis 2024; 45:247-261. [PMID: 38190483 DOI: 10.1093/carcin/bgae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/01/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024] Open
Abstract
We previously reported that RNF148 was involved in the ubiquitination-mediated degradation of CHAC2. However, its molecular mechanism was not determined. In this study, we investigated the role and mechanism of RNF148 in the progression of colorectal cancer (CRC), especially in the process of ubiquitination-mediated degradation of CHAC2. Our results revealed that RNF148 was upregulated in most CRC tissues, and its expression significantly correlated with the 3-year overall survival rate and most clinicopathological parameters of CRC patients. Furthermore, RNF148 served as an independent prognostic biomarker of CRC and promoted CRC cell proliferation and migration while inhibiting cell apoptosis and sensitivity to 5-FU. Mechanistically, RNF148 used its protease-associated domain to bind to the CHAC domain of CHAC2 and target it for degradation. In addition, we identified two phosphorylation and three ubiquitination residues of CHAC2 and identified Y118 and K102 as the critical phosphorylation and ubiquitination residues, respectively. We also identified CHAC2's and RNF148's interacting proteins and discovered their potential interaction network. In conclusion, our current study unveiled the role of RNF148 in CRC and the mechanism of RNF148 in the ubiquitination-mediated degradation of CHAC2, which shed light on providing potential prognostic biomarkers and molecular targets for CRC patients.
Collapse
Affiliation(s)
- Shuiping Liu
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China
- Division of Cellular and Molecular Research, Laboratory of Cancer Genomics, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Lvjia Zhuo
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Lu Chen
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ying He
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xudong Chen
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Hao Zhang
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yuan Zhou
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ziheng Ni
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shujuan Zhao
- Department of Respiratory Medicine of Affiliated Hospital, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiaotong Hu
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
5
|
Kannan MP, Sreeraman S, Somala CS, Kushwah RB, Mani SK, Sundaram V, Thirunavukarasou A. Advancement of targeted protein degradation strategies as therapeutics for undruggable disease targets. Future Med Chem 2023; 15:867-883. [PMID: 37254917 DOI: 10.4155/fmc-2023-0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/10/2023] [Indexed: 06/01/2023] Open
Abstract
Targeted protein degradation (TPD) aids in developing novel bifunctional small-molecule degraders and eliminates proteins of interest. The TPD approach shows promising results in oncological, neurogenerative, cardiovascular and gynecological drug development. We provide an overview of technology advancements in TPD, including molecular glues, proteolysis-targeting chimeras (PROTACs), lysosome-targeting chimeras, antibody-based PROTAC, GlueBody PROTAC, autophagy-targeting chimera, autophagosome-tethering compound, autophagy-targeting chimera and chaperone-mediated autophagy-based degraders. Here we discuss the development and evolution of the TPD field, the variety of proteins that PROTACs target and the biological repercussions of their degradation. We particularly highlight the recent improvements in TPD research that utilize autophagy or the endolysosomal pathway, which enables the targeting of undruggable targets.
Collapse
Affiliation(s)
- Mayuri P Kannan
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical & Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, 602105, India
- B-Aatral Biosciences Private Limited, Bangalore, Karnataka, 560091, India
| | - Sarojini Sreeraman
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical & Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, 602105, India
- SRIIC Lab, Sri Ramachandra Institute for Higher Education & Research, Chennai, Tamil Nadu, 600116, India
| | - Chaitanya S Somala
- B-Aatral Biosciences Private Limited, Bangalore, Karnataka, 560091, India
| | - Raja Bs Kushwah
- B-Aatral Biosciences Private Limited, Bangalore, Karnataka, 560091, India
- Department of Entomology and Agrilife Research, Texas A&M University, College Station, TX 77843, USA
| | - Saravanan K Mani
- B-Aatral Biosciences Private Limited, Bangalore, Karnataka, 560091, India
- Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, 600073, India
| | - Vickram Sundaram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical & Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, 602105, India
| | - Anand Thirunavukarasou
- B-Aatral Biosciences Private Limited, Bangalore, Karnataka, 560091, India
- SRIIC Lab, Sri Ramachandra Institute for Higher Education & Research, Chennai, Tamil Nadu, 600116, India
| |
Collapse
|
6
|
Huang G, Jiang Z, Zhu W, Wu Z. Exosomal circKDM4A Induces CUL4B to Promote Prostate Cancer Cell Malignancy in a miR-338-3p-Dependent Manner. Biochem Genet 2023; 61:390-409. [PMID: 35930171 DOI: 10.1007/s10528-022-10251-2] [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: 04/19/2022] [Accepted: 06/22/2022] [Indexed: 01/24/2023]
Abstract
Circular RNA lysine demethylase 4A (circKDM4A) is also named circ_0012098 and its abnormal expression has been confirmed in serum exosomes of prostate cancer (PC) patients. However, whether PC progression involves the exosomal circ_0012098 remains unknown. RNA expression of circKDM4A, microRNA-338-3p (miR-338-3p) and cullin 4B (CUL4B) was detected by quantitative real-time polymerase chain reaction. Protein expression was checked by Western blot. The positive expression rate of nuclear proliferation marker (ki-67) was analyzed by immunohistochemistry assay. Dual-luciferase reporter assay and RNA immunoprecipitation assay were used to identify the interaction between miR-338-3p and circKDM4A or CUL4B. Mouse model assay was performed to determine the effect of exosomal circKDM4A on tumorigenesis in vivo. CircKDM4A expression was significantly upregulated in the serum exosomes from PC patients compared with the exosomes from healthy volunteers. Exosomes treatment promoted the proliferation, migration and invasion of PC cells but inhibited apoptosis; however, these effects were attenuated after circKDM4A knockdown. Meanwhile, circKDM4A depletion restored exosome-increased circKDM4A expression. Additionally, circKDM4A acted as a miR-338-3p sponge, and miR-338-3p bound to CUL4B in PC cells. CircKDM4A regulated the effect of exosome-induced PC cell malignancy by interacting with miR-338-3p and CUL4B. Moreover, circKDM4A silencing relieved exosome-induced tumor growth in vivo. Exosomal circKDM4A promoted PC malignant progression by the miR-338-3p/CUL4B axis, providing a therapeutic target for PC.
Collapse
Affiliation(s)
- Guangyi Huang
- Department of Urology Surgery, the Fourth Affiliated Hospital Zhejiang University School of Medicine, Shangcheng Dadao, Yiwu City, 322001, Zhejiang Province, China.
| | - Zeping Jiang
- Department of Urology Surgery, the Fourth Affiliated Hospital Zhejiang University School of Medicine, Shangcheng Dadao, Yiwu City, 322001, Zhejiang Province, China
| | - Wuan Zhu
- Department of Urology Surgery, the Fourth Affiliated Hospital Zhejiang University School of Medicine, Shangcheng Dadao, Yiwu City, 322001, Zhejiang Province, China
| | - Zhiyue Wu
- Department of Urology Surgery, the Fourth Affiliated Hospital Zhejiang University School of Medicine, Shangcheng Dadao, Yiwu City, 322001, Zhejiang Province, China
| |
Collapse
|
7
|
Zhang X, Yang Y, Li D, Wu Z, Liu H, Zhao Z, Zhu H, Xie F, Li X. MOF negatively regulates estrogen receptor α signaling via CUL4B-mediated protein degradation in breast cancer. Front Oncol 2022; 12:868866. [PMID: 36212422 PMCID: PMC9539768 DOI: 10.3389/fonc.2022.868866] [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: 02/03/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Estrogen receptor α (ERα) is the dominant tumorigenesis driver in breast cancer (BC), and ERα-positive BC (ERα+ BC) accounts for more than two-thirds of BC cases. MOF (males absent on the first) is a highly conserved histone acetyltransferase that acetylates lysine 16 of histone H4 (H4K16) and several non-histone proteins. Unbalanced expression of MOF has been identified, and high MOF expression predicted a favorable prognosis in BC. However, the association of MOF with ERα and the regulatory mechanisms of MOF in ERα signaling remain elusive. Our study revealed that the expression of MOF is negatively correlated with that of ERα in BC. In ERα+ BC cells, MOF overexpression downregulated the protein abundance of ERα in both cytoplasm and nucleus, thus attenuating ERα-mediated transactivation as well as cellular proliferation and in vivo tumorigenicity of BC cells. MOF promoted ERα protein degradation through CUL4B-mediated ubiquitin–proteasome pathway and induced HSP90 hyperacetylation that led to the loss of chaperone protection of HSP90 to ERα. We also revealed that suppression of MOF restored ERα expression and increased the sensitivity of ERα-negative BC cells to tamoxifen treatment. These results provide a new insight into the tumor-suppressive role of MOF in BC via negatively regulating ERα action, suggesting that MOF might be a potential therapeutic target for BC.
Collapse
Affiliation(s)
- Xu Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yang Yang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Danyang Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- Rehabilitation Center, Qilu Hospital, Cheelo College of Medicine, Shandong University, Jinan, China
| | - Zhen Wu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Haoyu Liu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Ziyan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Hongying Zhu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Fei Xie
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiangzhi Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- *Correspondence: Xiangzhi Li,
| |
Collapse
|
8
|
Zhong M, Zhou L, Zou J, He Y, Fang Z, Xiang X. Cullin-4B promotes cell proliferation and invasion through inactivation of p53 signaling pathway in colorectal cancer. Pathol Res Pract 2021; 224:153520. [PMID: 34153655 DOI: 10.1016/j.prp.2021.153520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 06/06/2021] [Accepted: 06/06/2021] [Indexed: 12/24/2022]
Abstract
Cullin 4B (CUL4B) is a member of the Cullin RING E3 ligase family, which is found to be overexpressed in multiple cancers, thus facilitating tumorigenesis and progression. However, the correlation between CUL4B and p53 in colorectal cancer cells (CRC) remains to be further elucidated. In this study, we newly identified that CUL4B functions as a negative regulator of p53, thereby facilitating CRC tumorigenesis and progression. Our data has demonstrated that CUL4B was frequently overexpressed in CRC tissues, and its upregulation was closely correlated with disease progression and poor prognosis. Moreover, CUL4B knockdown suppressed cell proliferation, invasion and epithelial-mesenchymal transition (EMT) of CRC cells. Mechanistically, CUL4B depletion increased the expression of p53 protein and its downstream targets p21, PUMA and MDM2. Furthermore, CUL4B depletion prolonged the half-life of p53 protein, and CUL4B is a binding partner of MDM2. In conclusion, our study shed new lights on the complex regulatory network between CUL4B and p53, and clarifies this CUL4B-p53 axis contributes greatly to CRC tumorigenesis and progression.
Collapse
Affiliation(s)
- Min Zhong
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Ling Zhou
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Jianping Zou
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yan He
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Ziling Fang
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| | - Xiaojun Xiang
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
9
|
The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression. Int J Mol Sci 2021; 22:ijms22115754. [PMID: 34072267 PMCID: PMC8198665 DOI: 10.3390/ijms22115754] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023] Open
Abstract
The cell cycle is a collection of events by which cellular components such as genetic materials and cytoplasmic components are accurately divided into two daughter cells. The cell cycle transition is primarily driven by the activation of cyclin-dependent kinases (CDKs), which activities are regulated by the ubiquitin-mediated proteolysis of key regulators such as cyclins, CDK inhibitors (CKIs), other kinases and phosphatases. Thus, the ubiquitin-proteasome system (UPS) plays a pivotal role in the regulation of the cell cycle progression via recognition, interaction, and ubiquitination or deubiquitination of key proteins. The illegitimate degradation of tumor suppressor or abnormally high accumulation of oncoproteins often results in deregulation of cell proliferation, genomic instability, and cancer occurrence. In this review, we demonstrate the diversity and complexity of the regulation of UPS machinery of the cell cycle. A profound understanding of the ubiquitination machinery will provide new insights into the regulation of the cell cycle transition, cancer treatment, and the development of anti-cancer drugs.
Collapse
|
10
|
Ye L, Wang F, Wu H, Yang H, Yang Y, Ma Y, Xue A, Zhu J, Chen M, Wang J, Zhang QA. Functions and Targets of miR-335 in Cancer. Onco Targets Ther 2021; 14:3335-3349. [PMID: 34045870 PMCID: PMC8144171 DOI: 10.2147/ott.s305098] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/28/2021] [Indexed: 01/22/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs (18~25 nt in length) that act as master regulators of eukaryotic gene expression. They might play an oncogenic or tumor-suppressive role in multiple cancers. In recent decades, several studies have focused on the functions and mechanisms of miR-335 in cancer. The expression level of miR-335 in tissues and cells varies with cancer types, and miR-335 has been proposed as a potential biomarker for the prognosis of cancer. Besides, miR-335 may serve as an oncogene or tumor suppressor via regulating different targets or pathways in tumor initiation, development, and metastasis. Furthermore, miR-335 also influences tumor microenvironment and drug sensitivity. MiR-335 is regulated by various factors such as lncRNAs and microRNAs. In this review, we reveal the functions and targets of miR-335 in various cancers and its potential application as a possible biomarker in prognostic judgment and treatment of malignant tumors.
Collapse
Affiliation(s)
- Lingling Ye
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Fen Wang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Hao Wu
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Hui Yang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yan Yang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yajun Ma
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Aili Xue
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jing Zhu
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Meili Chen
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jinyan Wang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Quan An Zhang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
11
|
Ye X, Liu X, Gao M, Gong L, Tian F, Shen Y, Hu H, Sun G, Zou Y, Gong Y. CUL4B Promotes Temozolomide Resistance in Gliomas by Epigenetically Repressing CDNK1A Transcription. Front Oncol 2021; 11:638802. [PMID: 33869025 PMCID: PMC8050354 DOI: 10.3389/fonc.2021.638802] [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: 12/07/2020] [Accepted: 03/19/2021] [Indexed: 01/10/2023] Open
Abstract
Resistance to temozolomide (TMZ), the first-line chemotherapeutic drug for glioblastoma (GBM) and anaplastic gliomas, is one of the most significant obstacles in clinical treatment. TMZ resistance is regulated by complex genetic and epigenetic networks. Understanding the mechanisms of TMZ resistance can help to identify novel drug targets and more effective therapies. CUL4B has been shown to be upregulated and promotes progression and chemoresistance in several cancer types. However, its regulatory effect and mechanisms on TMZ resistance have not been elucidated. The aim of this study was to decipher the role and mechanism of CUL4B in TMZ resistance. Western blot and public datasets analysis showed that CUL4B was upregulated in glioma specimens. CUL4B elevation positively correlated with advanced pathological stage, tumor recurrence, malignant molecular subtype and poor survival in glioma patients receiving TMZ treatment. CUL4B expression was correlated with TMZ resistance in GBM cell lines. Knocking down CUL4B restored TMZ sensitivity, while upregulation of CUL4B promoted TMZ resistance in GBM cells. By employing senescence β-galactosidase staining, quantitative reverse transcription PCR and Chromatin immunoprecipitation experiments, we found that CUL4B coordinated histone deacetylase (HDAC) to co-occupy the CDKN1A promoter and epigenetically silenced CDKN1A transcription, leading to attenuation of TMZ-induced senescence and rendering the GBM cells TMZ resistance. Collectively, our findings identify a novel mechanism by which GBM cells develop resistance to TMZ and suggest that CUL4B inhibition may be beneficial for overcoming resistance.
Collapse
Affiliation(s)
- Xiang Ye
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaochen Liu
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Gao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Gong
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Tian
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yangli Shen
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huili Hu
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gongping Sun
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yongxin Zou
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
12
|
Hao Q, Chen Y, Zhou X. The Janus Face of p53-Targeting Ubiquitin Ligases. Cells 2020; 9:cells9071656. [PMID: 32660118 PMCID: PMC7407405 DOI: 10.3390/cells9071656] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
The tumor suppressor p53 prevents tumorigenesis and cancer progression by maintaining genomic stability and inducing cell growth arrest and apoptosis. Because of the extremely detrimental nature of wild-type p53, cancer cells usually mutate the TP53 gene in favor of their survival and propagation. Some of the mutant p53 proteins not only lose the wild-type activity, but also acquire oncogenic function, namely “gain-of-function”, to promote cancer development. Growing evidence has revealed that various E3 ubiquitin ligases are able to target both wild-type and mutant p53 for degradation or inactivation, and thus play divergent roles leading to cancer cell survival or death in the context of different p53 status. In this essay, we reviewed the recent progress in our understanding of the p53-targeting E3 ubiquitin ligases, and discussed the potential clinical implications of these E3 ubiquitin ligases in cancer therapy.
Collapse
Affiliation(s)
- Qian Hao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China;
| | - Yajie Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China;
| | - Xiang Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China;
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- Correspondence: ; Tel.: +86-21-54237325
| |
Collapse
|
13
|
Duan PJ, Zhao JH, Xie LL. Cul4B promotes the progression of ovarian cancer by upregulating the expression of CDK2 and CyclinD1. J Ovarian Res 2020; 13:76. [PMID: 32622365 PMCID: PMC7335446 DOI: 10.1186/s13048-020-00677-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background Ovarian cancer is one of the most common malignant tumors in the female reproductive system with the highest mortality rate. Cul4B participates in the oncogenesis and progression of several malignant tumors. However, the role of Cul4B in ovarian cancer has not been studied. Results High expression of intratumor Cul4B was associated with poor patient survival. Cul4B expression was associated with FIGO stage and Cul4B was independent risk factor of ovarian cancer disease-free survival and overall survival. In vitro studies revealed that overexpression of Cul4B promoted tumor proliferation while knockdown of Cul4B significantly inhibited the proliferation capacity of ovarian cancer cells. Mechanistically, Cul4B was found to promotes cell entering S phase from G0/G1 phase by regulating the expression of CDK2 and CyclinD1. Cul4B regulates the expression of CDK2 and CyclinD1 by repressing miR-372. Conclusions The results revealed that high expression of Cul4B is associated with poor ovarian cancer prognosis and Cul4B may serve as a potential treating target for an adjuvant therapy.
Collapse
Affiliation(s)
- Peng-Jing Duan
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Shandong Medical College, 80 Jintan Road, Linyi, 276000, Shandong, China
| | - Juan-Hong Zhao
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Shandong Medical College, 80 Jintan Road, Linyi, 276000, Shandong, China
| | - Li-Li Xie
- Department of Gynaecology, The people's hospital of Linshu, 182 West Shuhe Road, Linshu, 276700, Shandong, China.
| |
Collapse
|
14
|
Yu C, Ying J, Yu K, Shen W, Jiang M. Circ_0074027 Contributes to Nonsmall Cell Lung Cancer Progression by Upregulating CUL4B Expression Through miR-335-5p. Cancer Biother Radiopharm 2020; 37:73-83. [PMID: 32580576 DOI: 10.1089/cbr.2020.3579] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Circular RNAs (circRNAs) are crucial regulators in human cancers, including nonsmall cell lung cancer (NSCLC). In this study, we aim to explore the biological functions and molecular mechanisms of circ_0074027 in NSCLC. Methods: Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to determine the expression of circ_0074027, paired like homeodomain 1 (PITX1) mRNA, microRNA-335-5p (miR-335-5p), and cullin 4B (CUL4B) mRNA. The feature of circ_0074027 was analyzed by RNase R digestion assay. Flow cytometry analysis was adopted to analyze cell cycle and cell apoptosis. Cell counting kit-8 (CCK-8) assay and colony formation assay were performed to assess cell proliferation. Western blot assay was conducted to measure protein levels. Dual-luciferase reporter and RNA pull-down assays were carried out to verify the relationships among circ_0074027, miR-335-5p, and CUL4B. The murine xenograft model was established to investigate the role of circ_0074027 in vivo. Results: High expression of circ_0074027 was found in NSCLC tissues and cells. Circ_0074027 knockdown suppressed cell viability, cell cycle process, and colony formation and promoted apoptosis in NSCLC cells in vitro and inhibited tumor growth in vivo. Circ_0074027 acted as a sponge of miR-335-5p. The effect of circ_0074027 knockdown on NSCLC progression was weakened by miR-335-5p inhibition. Moreover, CUL4B was a target gene of miR-335-5p. CUL4B overexpression reversed the inhibitory effects on cell viability, cell cycle process, and colony formation and the promotional effect on cell apoptosis caused by miR-335-5p in NSCLC. Conclusion: Circ_0074027 facilitated NSCLC cell progression through regulating miR-335-5p/CUL4B axis.
Collapse
Affiliation(s)
- Chaoqun Yu
- Department of General Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Jianjian Ying
- Department of General Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Kaizhong Yu
- Department of General Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Weiyu Shen
- Department of General Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Maofen Jiang
- Department of Pathology, Beilun District People's Hospital, Ningbo, China
| |
Collapse
|
15
|
Yang X, Liu G, Li W, Zang L, Li D, Wang Q, Yu F, Xiang X. Silencing of zinc finger protein 703 inhibits medullary thyroid carcinoma cell proliferation in vitro and in vivo. Oncol Lett 2019; 19:943-951. [PMID: 31897207 PMCID: PMC6924197 DOI: 10.3892/ol.2019.11153] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 10/18/2019] [Indexed: 12/19/2022] Open
Abstract
Zinc finger protein 703 (ZNF703) is a new member of the zinc finger protein family of transcription factors that plays an important role during embryogenesis in metazoans. The overexpression of ZNF703 contributes to tumorigenesis and progression of a number of malignancies by activating the Akt/mammalian target of rapamycin (mTOR) signaling pathway. This pathway is activated in medullary thyroid cancer (MTC), but its mechanism of action is not yet fully understood. The aim of the present study was to examine the role of ZNF703 and its association with Akt/mTOR activation in MTC. The present study used the phosphorylation of Akt1 protein at serine 473 (pAkt473) as an indicator of signaling activation. Immunohistochemistry (IHC) staining and western blot analyses were performed in order to examine the expression of ZNF703 in 34 cases of MTC and 12 cases of corresponding normal thyroid tissues. ZNF703 expression in MTC was significantly higher compared with the corresponding normal thyroid tissues (P<0.05). Furthermore, expression of ZNF703 was associated with tumor size, lymph node metastasis and advanced stage of disease. IHC also demonstrated that the level of ZNF703 was positively correlated with p-Akt473 in the 34 cases of MTC. The human MTC cell line TT was selected for further investigation as TT cells exhibit Akt/mTOR activation. The biological effects of silencing ZNF703 in TT cells on proliferation and apoptosis, both in vitro and in vivo were investigated in the present study. ZNF703 silencing inhibited the proliferation of TT cells in vitro and inhibited xenograft tumor growth in vivo. These effects were accompanied by the substantial decrease of pAkt473 and the induction of p53 protein. These results demonstrate that ZNF703 may play a relevant role in MTC due to its association with the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaolin Yang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Geling Liu
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Weijuan Li
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Luyang Zang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Ding Li
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Qianqian Wang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Fang Yu
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Xiuxiu Xiang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
16
|
Zhang H, Wang A, Tan Y, Wang S, Ma Q, Chen X, He Z. NCBP1 promotes the development of lung adenocarcinoma through up-regulation of CUL4B. J Cell Mol Med 2019; 23:6965-6977. [PMID: 31448526 PMCID: PMC6787490 DOI: 10.1111/jcmm.14581] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 06/06/2019] [Accepted: 06/24/2019] [Indexed: 01/01/2023] Open
Abstract
Lung cancer is the most frequent cancer type and is the leading cause of tumour‐associated deaths worldwide. Nuclear cap‐binding protein 1 (NCBP1) is necessary for capped RNA processing and intracellular localization. It has been reported that silencing of NCBP1 resulted in cell growth reduction in HeLa cells. Nevertheless, its clinical significance and underlying molecular mechanisms in non–small‐cell lung cancer remain unclear. In this study, we found that NCBP1 was significantly overexpressed in lung cancer tissues and several lung cancer cell lines. Through knockdown and overexpression experiments, we showed that NCBP1 promoted lung cancer cell growth, wound healing ability, migration and epithelial‐mesenchymal transition. Mechanistically, we found that cullin 4B (CUL4B) was a downstream target gene of NCBP1 in NSCLC. NCBP1 up‐regulated CUL4B expression via interaction with nuclear cap‐binding protein 3 (NCBP3). CUL4B silencing significantly reversed NCBP1‐induced tumorigenesis in vitro. Based on these findings, we propose a model involving the NCBP1‐NCBP3‐CUL4B oncoprotein axis, providing novel insight into how CUL4B is activated and contributes to LUAD progression.
Collapse
Affiliation(s)
- Huijun Zhang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - An Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Yulong Tan
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Shaohua Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Qinyun Ma
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Xiaofeng Chen
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Zelai He
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
17
|
Zhao M, Qi M, Li X, Hu J, Zhang J, Jiao M, Bai X, Peng X, Han B. CUL4B/miR-33b/C-MYC axis promotes prostate cancer progression. Prostate 2019; 79:480-488. [PMID: 30609075 DOI: 10.1002/pros.23754] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/27/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Cullin 4B (CUL4B), a scaffold protein that assembles CRL4B ubiquitin ligase complexes, is overexpressed in many types of solid tumors and contributes to epigenetic silencing of tumor suppressors. However, its clinical significance and underlying molecular mechanisms in prostate cancer (PCa) remain unknown. METHODS The clinical significance of CUL4B in PCa was characterized by in silico method. RT-qPCR and Western blot were used to study the transcript and protein expression levels of CUL4B and C-MYC. Bioinformatics tools, chromatin immunoprecipitation (ChIP) and luciferase reporter assay were utilized to identify and characterize the microRNAs (miRNAs) regulated by CUL4B. The biological function of CUL4B and miR-33b-5p was evaluated by MTS, transwell, and wound healing assays, accordingly. RESULTS CUL4B is significantly overexpressed in PCa tissues compared with benign prostatic tissues and its overexpression is correlated with poor prognosis. CUL4B promotes proliferation and aggressiveness of PCa cells in vitro. Mechanistically, we demonstrate that CUL4B upregulates the expression of C-MYC at post-transcriptional level through epigenetic silencing of miR-33b-5p. Importantly, CUL4B-induced oncogenic activity in PCa by targeting C-MYC is repressed by miR-33b-5p. CONCLUSIONS Our results suggested a novel CUL4B/miR-33b/C-MYC axis implicated in PCa cell growth and progression. This might provide novel insight into how CUL4B contributed to PCa aggressiveness and progression.
Collapse
Affiliation(s)
- Mingfeng Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
- Department of Pathology, Binzhou Medical University, Binzhou, China
| | - Mei Qi
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
| | - Xinjun Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
- Department of Pathology, Binzhou People's Hospital, Binzhou, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Meng Jiao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
| | - Xinnuo Bai
- Department of Public Health Sciences, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Xijia Peng
- Human Biology Program, University of Toronto, Toronto, Ontario, Canada
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
- Department of Pathology, Shandong University QiLu Hospital, Jinan, China
| |
Collapse
|
18
|
Gupta I, Singh K, Varshney NK, Khan S. Delineating Crosstalk Mechanisms of the Ubiquitin Proteasome System That Regulate Apoptosis. Front Cell Dev Biol 2018; 6:11. [PMID: 29479529 PMCID: PMC5811474 DOI: 10.3389/fcell.2018.00011] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/26/2018] [Indexed: 01/10/2023] Open
Abstract
Regulatory functions of the ubiquitin-proteasome system (UPS) are exercised mainly by the ubiquitin ligases and deubiquitinating enzymes. Degradation of apoptotic proteins by UPS is central to the maintenance of cell health, and deregulation of this process is associated with several diseases including tumors, neurodegenerative disorders, diabetes, and inflammation. Therefore, it is the view that interrogating protein turnover in cells can offer a strategy for delineating disease-causing mechanistic perturbations and facilitate identification of drug targets. In this review, we are summarizing an overview to elucidate the updated knowledge on the molecular interplay between the apoptosis and UPS pathways. We have condensed around 100 enzymes of UPS machinery from the literature that ubiquitinates or deubiquitinates the apoptotic proteins and regulates the cell fate. We have also provided a detailed insight into how the UPS proteins are able to fine-tune the intrinsic, extrinsic, and p53-mediated apoptotic pathways to regulate cell survival or cell death. This review provides a comprehensive overview of the potential of UPS players as a drug target for cancer and other human disorders.
Collapse
Affiliation(s)
- Ishita Gupta
- Structural Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,Drug Discovery Research Centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Kanika Singh
- Drug Discovery Research Centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Nishant K Varshney
- Drug Discovery Research Centre, Translational Health Science and Technology Institute, Faridabad, India
| | - Sameena Khan
- Drug Discovery Research Centre, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
19
|
p53 stability is regulated by diverse deubiquitinating enzymes. Biochim Biophys Acta Rev Cancer 2017; 1868:404-411. [PMID: 28801249 DOI: 10.1016/j.bbcan.2017.08.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/21/2017] [Accepted: 08/05/2017] [Indexed: 02/01/2023]
Abstract
The tumor suppressor protein p53 has a variety of roles in responses to various stress signals. In such responses, p53 activates specific transcriptional targets that control cell cycle arrest, DNA repair, angiogenesis, autophagy, metabolism, migration, aging, senescence, and apoptosis. Since p53 has been identified as the most frequently altered gene in human cancers, regulation and stabilization of its normal functions are important. Stability of p53 is regulated by the ubiquitin-proteasome pathway (UPP). Furthermore, it is readjusted by deubiquitination via deubiquitinating enzymes (DUBs) that can eliminate ubiquitin from p53. Diverse DUBs directly or indirectly affect the ubiquitination of p53 and, consequently, regulate various cellular processes associated with p53. As maintenance of p53 is regulated by a variety of DUBs, the interaction of DUBs and p53 can affect diseases such as cancer. Currently, DUBs have a central role in our understanding of various cancers, and some have potential in the development of effective therapeutic strategies. This review summarizes the current knowledge of p53 and of the interconnection between p53 and DUBs.
Collapse
|
20
|
miR-181a-2 downregulates the E3 ubiquitin ligase CUL4A transcript and promotes cell proliferation. Med Oncol 2017; 34:146. [PMID: 28730334 DOI: 10.1007/s12032-017-1006-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 07/15/2017] [Indexed: 10/25/2022]
Abstract
MiR-181a-2 plays a major role in cell proliferation both positively and negatively depending on tissue type by targeting several regulators 3'UTR regions. We have predicted several targets for miR-181a-2 through computational approaches and characterized one its interesting target, CUL4A, an E3 ubiquitin ligase. CUL4A regulates diverse functions in the cells including DNA repair, DNA replication, cell cycle, genomic stability through polyubiquitination of target proteins. Deregulation of both miR-181a-2 and CUL4A are reported in many cancerous cells, but the functional link between them is unknown. We show that miR-181a-5p binds to 3'UTR of CUL4A and regulates its transcripts levels in HEK293 cells through overexpression studies. In addition, by using MTT and Neutral red assays, we showed that miR-181a-2 overexpression increased the proliferation in HEK293 cells. Moreover, cell cycle analysis using flow cytometer revealed that an increase in S-phase cells upon the overexpression of miR-181a-2. Though several miRNAs are known to downregulate the CUL4A levels, here we show that miR-181a-2 also participates in the downregulation of CUL4A. Taken together, our data demonstrated that miR-181a-2 increases the cell proliferation in HEK293 cells possibly through the downregulation of CUL4A.
Collapse
|
21
|
Yuan J, Jiang B, Zhang A, Qian Y, Tan H, Gao J, Shao C, Gong Y. Accelerated hepatocellular carcinoma development in CUL4B transgenic mice. Oncotarget 2016; 6:15209-21. [PMID: 25945838 PMCID: PMC4558146 DOI: 10.18632/oncotarget.3829] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/26/2015] [Indexed: 01/08/2023] Open
Abstract
Cullin 4B (CUL4B) is a component of the Cullin 4B-Ring E3 ligase (CRL4B) complex that functions in proteolysis and in epigenetic regulation. CUL4B possesses tumor-promoting properties and is markedly upregulated in many types of human cancers. To determine the role of CUL4B in liver tumorigenesis, we generated transgenic mice that expressed human CUL4B in livers and other tissues and evaluated the development of spontaneous and chemically-induced hepatocellular carcinomas. We observed that CUL4B transgenic mice spontaneously developed liver tumors at a high incidence at old ages and exhibited enhanced DEN-induced hepatocarcinogenesis. There was a high proliferation rate in the livers of CUL4B transgenic mice that was accompanied by increased levels of Cdk1, Cdk4 and cyclin D1 and decreased level of p16. The transgenic mice also exhibited increased compensatory proliferation after DEN-induced liver injury, which was accompanied by activation of Akt, Erk, p38 and NF-κB. We also found that Prdx3 was downregulated and that DEN induced a higher level of reactive oxygen species in the livers of transgenic mice. Together, our results demonstrate a critical role of CUL4B in hepatocarcinogenesis in mice.
Collapse
Affiliation(s)
- Jupeng Yuan
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, China
| | - Baichun Jiang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, China
| | - Aizhen Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Life Science, Jinan, China
| | - Yanyan Qian
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, China
| | - Haining Tan
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, China
| | - Jiangang Gao
- Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Life Science, Jinan, China
| | - Changshun Shao
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
22
|
Chen P, Yao GD. The role of cullin proteins in gastric cancer. Tumour Biol 2015; 37:29-37. [PMID: 26472722 DOI: 10.1007/s13277-015-4154-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/23/2015] [Indexed: 01/09/2023] Open
Abstract
The cullin proteins are a family of scaffolding proteins that associate with RING proteins and ubiquitin E3 ligases and mediate substrate-receptor bindings. Thus, cullin proteins regulate the specificity of ubiquitin targeting in the regulation of proteins involved in various cellular processes, including proliferation, differentiation, and apoptosis. There are seven cullin proteins that have been identified in eukaryotes: CUL1, CUL2, CUL3, CUL4A, CUL4B, CUL5, and CUL7/p53-associated parkin-like cytoplasmic protein. All of these proteins contain a conserved cullin homology domain that binds to RING box proteins. Cullin-RING ubiquitin ligase complexes are activated upon post-translational modification by neural precursor cell-expressed, developmentally downregulated protein 8. The aberrant expression of several cullin proteins has been implicated in many cancers though the significance in gastric cancer has been less well investigated. This review provides the first systematic discussion of the associations between all members of the cullin protein family and gastric cancer. Functional and regulatory mechanisms of cullin proteins in gastric carcinoma progression are also summarized along with a discussion concerning future research areas. Accumulating evidence suggests a critical role of cullin proteins in tumorigenesis, and a better understanding of the function of these individual cullin proteins and their targets will help identify potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Peng Chen
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Tong-Dao-Bei Street, Hohhot, Inner Mongolia, 010050, People's Republic of China
| | - Guo-Dong Yao
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Tong-Dao-Bei Street, Hohhot, Inner Mongolia, 010050, People's Republic of China.
| |
Collapse
|
23
|
Hannah J, Zhou P. Distinct and overlapping functions of the cullin E3 ligase scaffolding proteins CUL4A and CUL4B. Gene 2015; 573:33-45. [PMID: 26344709 DOI: 10.1016/j.gene.2015.08.064] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/03/2015] [Accepted: 08/27/2015] [Indexed: 01/29/2023]
Abstract
The cullin 4 subfamily of genes includes CUL4A and CUL4B, which share a mostly identical amino acid sequence aside from the elongated N-terminal region in CUL4B. Both act as scaffolding proteins for modular cullin RING ligase 4 (CRL4) complexes which promote the ubiquitination of a variety of substrates. CRL4 function is vital to cells as loss of both genes or their shared substrate adaptor protein DDB1 halts proliferation and eventually leads to cell death. Due to their high structural similarity, CUL4A and CUL4B share a substantial overlap in function. However, in some cases, differences in subcellular localization, spatiotemporal expression patterns and stress-inducibility preclude functional compensation. In this review, we highlight the most essential functions of the CUL4 genes in: DNA repair and replication, chromatin-remodeling, cell cycle regulation, embryogenesis, hematopoiesis and spermatogenesis. CUL4 genes are also clinically relevant as dysregulation can contribute to the onset of cancer and CRL4 complexes are often hijacked by certain viruses to promote viral replication and survival. Also, mutations in CUL4B have been implicated in a subset of patients suffering from syndromic X-linked intellectual disability (AKA mental retardation). Interestingly, the antitumor effects of immunomodulatory drugs are caused by their binding to the CRL4CRBN complex and re-directing the E3 ligase towards the Ikaros transcription factors IKZF1 and IKZF3. Because of their influence over key cellular functions and relevance to human disease, CRL4s are considered promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jeffrey Hannah
- Department of Pathology, Weill Cornell Medical College, 1300 York Ave. NY, NY 10065, United States.
| | - Pengbo Zhou
- Department of Pathology, Weill Cornell Medical College, 1300 York Ave. NY, NY 10065, United States.
| |
Collapse
|
24
|
Ke JY, Dai CJ, Wu WL, Gao JH, Xia AJ, Liu GP, Lv KS, Wu CL. USP11 regulates p53 stability by deubiquitinating p53. J Zhejiang Univ Sci B 2015; 15:1032-8. [PMID: 25471832 DOI: 10.1631/jzus.b1400180] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The p53 tumor suppressor protein coordinates the cellular responses to a broad range of cellular stresses, leading to DNA repair, cell cycle arrest or apoptosis. The stability of p53 is essential for its tumor suppressor function, which is tightly controlled by ubiquitin-dependent degradation primarily through its negative regulator murine double minute 2 (Mdm2). To better understand the regulation of p53, we tested the interaction between p53 and USP11 using co-immunoprecipitation. The results show that USP11, an ubiquitin-specific protease, forms specific complexes with p53 and stabilizes p53 by deubiquitinating it. Moreover, down-regulation of USP11 dramatically attenuated p53 induction in response to DNA damage stress. These findings reveal that USP11 is a novel regulator of p53, which is required for p53 activation in response to DNA damage.
Collapse
Affiliation(s)
- Jia-ying Ke
- College of Chemistry and Life Science, Quanzhou Normal University, Quanzhou 36200, China; The Higher Educational Key Laboratory for Molecular Biology and Pharmacology of Fujian Province, Quanzhou 36200, China; Xiamen Women and Children Health Hospital, Xiamen 361005, China; Shouguang People's Hospital, Shouguang 262700, China; Department of Orthopedics, Central Hospital of Zibo, Zibo 255000, China; Department of Pathology, University of Chicago, Chicago 60102, Illinois, USA; Department of Pathology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 36200, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhao W, Jiang B, Hu H, Zhang S, Lv S, Yuan J, Qian Y, Zou Y, Li X, Jiang H, Liu F, Shao C, Gong Y. Lack of CUL4B leads to increased abundance of GFAP-positive cells that is mediated by PTGDS in mouse brain. Hum Mol Genet 2015; 24:4686-97. [PMID: 26025376 DOI: 10.1093/hmg/ddv200] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 05/26/2015] [Indexed: 01/05/2023] Open
Abstract
Astrocytes are the most abundant cell type in the mammalian brain and are important for the functions of the central nervous system. Glial fibrillary acidic protein (GFAP) is regarded as a hallmark of mature astrocytes, though some GFPA-positive cells may act as neural stem cells. Missense heterozygous mutations in GFAP cause Alexander disease that manifests leukodystrophy and intellectual disability. Here, we show that CUL4B, a scaffold protein that assembles E3 ubiquitin ligase, represses the expression of GFAP in neural progenitor cells (NPCs) during brain development. Lack of Cul4b in NPCs in cultures led to increased generation of astrocytes, marked by GFAP and S100β. The GFAP+ cells were also found to be more abundant in the brains of nervous system-specific Cul4b knockout mice in vivo. Moreover, we demonstrated that the increased generation of GFAP+ cells from Cul4b-null NPCs was mediated by an upregulation of prostaglandin D2 synthase PTGDS. We showed that the increased GFAP expression can be attenuated by pharmacological inhibition of the PTGDS enzymatic activity or by shRNA-mediated knockdown of Ptgds. Importantly, exogenously added PTGDS could promote the generation of GFAP+ cells from wild-type NPCs. We further observed that Ptgds is targeted and repressed by the CUL4B/PRC2 complex. Together, our results demonstrate CUL4B as a negative regulator of GFAP expression during neural development.
Collapse
Affiliation(s)
- Wei Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuqian Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuaishuai Lv
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Jupeng Yuan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yanyan Qian
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xi Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Hong Jiang
- Institute of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong 250012, China and
| | - Fang Liu
- Department of Neuroscience, Centre for Addiction and Mental Health, Toronto, Ontario, Canada M5T 1R8
| | - Changshun Shao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China,
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China,
| |
Collapse
|
26
|
Brinkmann K, Schell M, Hoppe T, Kashkar H. Regulation of the DNA damage response by ubiquitin conjugation. Front Genet 2015; 6:98. [PMID: 25806049 PMCID: PMC4354423 DOI: 10.3389/fgene.2015.00098] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/23/2015] [Indexed: 12/12/2022] Open
Abstract
In response to DNA damage, cells activate a highly conserved and complex kinase-based signaling network, commonly referred to as the DNA damage response (DDR), to safeguard genomic integrity. The DDR consists of a set of tightly regulated events, including detection of DNA damage, accumulation of DNA repair factors at the site of damage, and finally physical repair of the lesion. Upon overwhelming damage the DDR provokes detrimental cellular actions by involving the apoptotic machinery and inducing a coordinated demise of the damaged cells (DNA damage-induced apoptosis, DDIA). These diverse actions involve transcriptional activation of several genes that govern the DDR. Moreover, recent observations highlighted the role of ubiquitylation in orchestrating the DDR, providing a dynamic cellular regulatory circuit helping to guarantee genomic stability and cellular homeostasis (Popovic et al., 2014). One of the hallmarks of human cancer is genomic instability (Hanahan and Weinberg, 2011). Not surprisingly, deregulation of the DDR can lead to human diseases, including cancer, and can induce resistance to genotoxic anti-cancer therapy (Lord and Ashworth, 2012). Here, we summarize the role of ubiquitin-signaling in the DDR with special emphasis on its role in cancer and highlight the therapeutic value of the ubiquitin-conjugation machinery as a target in anti-cancer treatment strategy.
Collapse
Affiliation(s)
- Kerstin Brinkmann
- Centre for Molecular Medicine Cologne and Institute for Medical Microbiology, Immunology and Hygiene, University Hospital of CologneCologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital of CologneCologne, Germany
| | - Michael Schell
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital of CologneCologne, Germany
- Institute for Genetics, University of CologneCologne, Germany
| | - Thorsten Hoppe
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital of CologneCologne, Germany
- Institute for Genetics, University of CologneCologne, Germany
| | - Hamid Kashkar
- Centre for Molecular Medicine Cologne and Institute for Medical Microbiology, Immunology and Hygiene, University Hospital of CologneCologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital of CologneCologne, Germany
| |
Collapse
|
27
|
Abidi N, Xirodimas DP. Regulation of cancer-related pathways by protein NEDDylation and strategies for the use of NEDD8 inhibitors in the clinic. Endocr Relat Cancer 2015; 22:T55-70. [PMID: 25504797 DOI: 10.1530/erc-14-0315] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Post-translational modification of proteins with ubiquitin and ubiquitin-like molecules (UBLs) controls a vast if not every biological process in the cell. It is not surprising that deregulation in ubiquitin and UBL signalling has been implicated in the pathogenesis of many diseases and that these pathways are considered as major targets for therapeutic intervention. In this review, we summarise recent advances in our understanding of the role of the UBL neural precursor cell expressed developmentally downregulated-8 (NEDD8) in cancer-related processes and potential strategies for the use of NEDD8 inhibitors as chemotherapeutics.
Collapse
Affiliation(s)
- Naima Abidi
- Centre de Recherche de Biochimie MacromoléculaireUMR5235, 1919 Route de Mende, Montpellier 34293, France
| | - Dimitris P Xirodimas
- Centre de Recherche de Biochimie MacromoléculaireUMR5235, 1919 Route de Mende, Montpellier 34293, France
| |
Collapse
|
28
|
Wei Z, Guo H, Liu Z, Zhang X, Liu Q, Qian Y, Gong Y, Shao C. CUL4B impedes stress-induced cellular senescence by dampening a p53-reactive oxygen species positive feedback loop. Free Radic Biol Med 2015; 79:1-13. [PMID: 25464270 DOI: 10.1016/j.freeradbiomed.2014.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/05/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
Abstract
Tumor suppressor p53 is known to regulate the level of intracellular reactive oxygen species (ROS). It can either alleviate oxidative stress under physiological and mildly stressed conditions or exacerbate oxidative stress under highly stressed conditions. We here report that a p53-ROS positive feedback loop drives a senescence program in normal human fibroblasts (NHFs) and this senescence-driving loop is negatively regulated by CUL4B. CUL4B, which can assemble various ubiquitin E3 ligases, was found to be downregulated in stress-induced senescent cells, but not in replicative senescent cells. We observed that p53-dependent ROS production was significantly augmented and stress-induced senescence was greatly enhanced when CUL4B was absent or depleted. Ectopic expression of CUL4B, on the other hand, blunted p53 activation, reduced ROS production, and attenuated cellular senescence in cells treated with H2O2. CUL4B was shown to promote p53 ubiquitination and proteosomal degradation in NHFs exposed to oxidative stress, thus dampening the p53-dependent cellular senescence. Together, our results established a critical role of CUL4B in negatively regulating the p53-ROS positive feedback loop that drives cellular senescence.
Collapse
Affiliation(s)
- Zhao Wei
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Haiyang Guo
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Zhaojian Liu
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xiyu Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Qiao Liu
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yanyan Qian
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Changshun Shao
- Key Laboratory of Experimental Teratology, Ministry of Education/Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China; Department of Genetics/Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
29
|
Chen Z, Sui J, Zhang F, Zhang C. Cullin family proteins and tumorigenesis: genetic association and molecular mechanisms. J Cancer 2015; 6:233-42. [PMID: 25663940 PMCID: PMC4317758 DOI: 10.7150/jca.11076] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/08/2014] [Indexed: 12/13/2022] Open
Abstract
Cullin family proteins function as scaffolds to form numerous E3 ubiquitin ligases with RING proteins, adaptor proteins and substrate recognition receptors. These E3 ligases further recognize numerous substrates to participate in a variety of cellular processes, such as DNA damage and repair, cell death and cell cycle progression. Clinically, cullin-associated E3 ligases have been identified to involve numerous human diseases, especially with regard to multiple cancer types. Over the past few years, our understanding of cullin proteins and their functions in genome stability and tumorigenesis has expanded enormously. Herein, this review briefly provides current perspectives on cullin protein functions, and mainly summarizes and discusses molecular mechanisms of cullin proteins in tumorigenesis.
Collapse
Affiliation(s)
- Zhi Chen
- 1. Orthopedics Department, Changhai Hospital Affiliated to Second Military Medical University, Shanghai, China, 200433
| | - Jie Sui
- 2. Orthopedics Department, 102 Hospital of People's Liberation Army, Changzhou, Jiangsu, China, 213003
| | - Fan Zhang
- 1. Orthopedics Department, Changhai Hospital Affiliated to Second Military Medical University, Shanghai, China, 200433
| | - Caiguo Zhang
- 3. Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA, 80045
| |
Collapse
|
30
|
Thirunavukarasou A, Govindarajalu G, Singh P, Bandi V, Muthu K, Baluchamy S. Cullin 4A and 4B ubiquitin ligases interact with γ-tubulin and induce its polyubiquitination. Mol Cell Biochem 2014; 401:219-28. [DOI: 10.1007/s11010-014-2309-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 12/20/2014] [Indexed: 11/27/2022]
|