1
|
Guo Q, Qin H, Chen Z, Zhang W, Zheng L, Qin T. Key roles of ubiquitination in regulating critical regulators of cancer stem cell functionality. Genes Dis 2025; 12:101311. [PMID: 40034124 PMCID: PMC11875185 DOI: 10.1016/j.gendis.2024.101311] [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: 11/02/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 03/05/2025] Open
Abstract
The ubiquitin (Ub) system, a ubiquitous presence across eukaryotes, plays a crucial role in the precise orchestration of diverse cellular protein processes. From steering cellular signaling pathways and orchestrating cell cycle progression to guiding receptor trafficking and modulating immune responses, this process plays a crucial role in regulating various biological functions. The dysregulation of Ub-mediated signaling pathways in prevalent cancers ushers in a spectrum of clinical outcomes ranging from tumorigenesis and metastasis to recurrence and drug resistance. Ubiquitination, a linchpin process mediated by Ub, assumes a central mantle in molding cellular signaling dynamics. It navigates transitions in biological cues and ultimately shapes the destiny of proteins. Recent years have witnessed an upsurge in the momentum surrounding the development of protein-based therapeutics aimed at targeting the Ub system under the sway of cancer stem cells. The article provides a comprehensive overview of the ongoing in-depth discussions regarding the regulation of the Ub system and its impact on the development of cancer stem cells. Amidst the tapestry of insights, the article delves into the expansive roles of E3 Ub ligases, deubiquitinases, and transcription factors entwined with cancer stem cells. Furthermore, the spotlight turns to the interplay with pivotal signaling pathways the Notch, Hedgehog, Wnt/β-catenin, and Hippo-YAP signaling pathways all play crucial roles in the regulation of cancer stem cells followed by the specific modulation of Ub-proteasome.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou 550014, China
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Artificial Intelligence and IoT Smart Medical Engineering Research Center of Henan Province, Zhengzhou, Henan 450008, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Tingting Qin
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| |
Collapse
|
2
|
You R, Li Y, Jiang Y, Hu D, Gu M, Zhou W, Zhang S, Bai M, Yang Y, Zhang Y, Huang S, Jia Z, Zhang A. WWP2 deletion aggravates acute kidney injury by targeting CDC20/autophagy axis. J Adv Res 2025; 71:471-485. [PMID: 38909885 DOI: 10.1016/j.jare.2024.06.015] [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: 03/30/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) is associated with high morbidity and mortality rates. The molecular mechanisms underlying AKI are currently being extensively investigated. WWP2 is an E3 ligase that regulates cell proliferation and differentiation. Whether WWP2 plays a regulatory role in AKI remains to be elucidated. OBJECTIVES We aimed to investigate the implication of WWP2 in AKI and its underlying mechanism in the present study. METHODS We utilized renal tissues from patients with AKI and established AKI models in global or tubule-specific knockout (cKO) mice strains to study WWP2's implication in AKI. We also systemically analyzed ubiquitylation omics and proteomics to decipher the underlying mechanism. RESULTS In the present study, we found that WWP2 expression significantly increased in the tubules of kidneys with AKI. Global or tubule-specific knockout of WWP2 significantly aggravated renal dysfunction and tubular injury in AKI kidneys, whereas WWP2 overexpression significantly protected tubular epithelial cells against cisplatin. WWP2 deficiency profoundly affected autophagy in AKI kidneys. Further analysis with ubiquitylation omics, quantitative proteomics and experimental validation suggested that WWP2 mediated poly-ubiquitylation of CDC20, a negative regulator of autophagy. CDC20 was significantly decreased in AKI kidneys, and selective inhibiting CDC20 with apcin profoundly alleviated renal dysfunction and tubular injury in the cisplatin model with or without WWP2 cKO, indicating that CDC20 may serve as a downstream target of WWP2 in AKI. Inhibiting autophagy with 3-methyladenine blocked apcin's protection against cisplatin-induced renal tubular cell injury. Activating autophagy by rapamycin significantly protected against cisplatin-induced AKI in WWP2 cKO mice, whereas inhibiting autophagy by 3-methyladenine further aggravated apoptosis in cisplatin-exposed WWP2 KO cells. CONCLUSION Taken together, our data indicated that the WWP2/CDC20/autophagy may be an essential intrinsic protective mechanism against AKI. Further activating WWP2 or inhibiting CDC20 may be novel therapeutic strategies for AKI.
Collapse
Affiliation(s)
- Ran You
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yanwei Li
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China; School of Medicine, Southeast University, Nanjing, China
| | - Yuteng Jiang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; School of Medicine, Southeast University, Nanjing, China
| | - Dandan Hu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Menglei Gu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mi Bai
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yunwen Yang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China; School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
3
|
Datta RR, Akdogan D, Tezcan EB, Onal P. Versatile roles of disordered transcription factor effector domains in transcriptional regulation. FEBS J 2025. [PMID: 39888268 DOI: 10.1111/febs.17424] [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: 09/01/2024] [Revised: 11/25/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Transcription, a crucial step in the regulation of gene expression, is tightly controlled and involves several essential processes, such as chromatin organization, recognition of the specific genomic sequences, DNA binding, and ultimately recruiting the transcriptional machinery to facilitate transcript synthesis. At the center of this regulation are transcription factors (TFs), which comprise at least one DNA-binding domain (DBD) and an effector domain (ED). Although the structure and function of DBDs have been well studied, our knowledge of the structure and function of effector domains is limited. EDs are of particular importance in generating distinct transcriptional responses between protein members of the same TF family that have similar DBDs and specificities. The study of transcriptional activity conferred by effector domains has traditionally been conducted through examining protein-protein interactions. However, recent research has uncovered alternative mechanisms by which EDs regulate gene expression, such as the formation of condensates that increase the local concentration of transcription factors, cofactors, and coregulated genes, as well as DNA binding. Here, we provide a comprehensive overview of the known roles of transcription factor EDs, with a specific focus on disordered regions. Additionally, we emphasize the significance of intrinsically disordered regions (IDRs) during transcriptional regulation. We examine the mechanisms underlying the establishment and maintenance of transcriptional specificity through the structural properties of predominantly disordered EDs. We then provide a comprehensive overview of the current understanding of these domains, including their physical and chemical characteristics, as well as their functional roles.
Collapse
Affiliation(s)
| | - Dilan Akdogan
- Molecular Biology and Genetics Department, Ihsan Dogramaci Bilkent University, Ankara, Turkey
| | - Elif B Tezcan
- Molecular Biology and Genetics Department, Ihsan Dogramaci Bilkent University, Ankara, Turkey
| | - Pinar Onal
- Molecular Biology and Genetics Department, Ihsan Dogramaci Bilkent University, Ankara, Turkey
| |
Collapse
|
4
|
Huang C, Jiang T, Pan W, Feng T, Zhou X, Wu Q, Ma F, Dai J. Ubiquitination of NS1 Confers Differential Adaptation of Zika Virus in Mammalian Hosts and Mosquito Vectors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408024. [PMID: 39159062 PMCID: PMC11497017 DOI: 10.1002/advs.202408024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Indexed: 08/21/2024]
Abstract
Arboviruses, transmitted by medical arthropods, pose a serious health threat worldwide. During viral infection, Post Translational Modifications (PTMs) are present on both host and viral proteins, regulating multiple processes of the viral lifecycle. In this study, a mammalian E3 ubiquitin ligase WWP2 (WW domain containing E3 ubiquitin ligase 2) is identified, which interacts with the NS1 protein of Zika virus (ZIKV) and mediates K63 and K48 ubiquitination of Lys 265 and Lys 284, respectively. WWP2-mediated NS1 ubiquitination leads to NS1 degradation via the ubiquitin-proteasome pathway, thereby inhibiting ZIKV infection in mammalian hosts. Simultaneously, it is found Su(dx), a protein highly homologous to host WWP2 in mosquitoes, is capable of ubiquitinating NS1 in mosquito cells. Unexpectedly, ubiquitination of NS1 in mosquitoes does not lead to NS1 degradation; instead, it promotes viral infection in mosquitoes. Correspondingly, the NS1 K265R mutant virus is less infectious to mosquitoes than the wild-type (WT) virus. The above results suggest that the ubiquitination of the NS1 protein confers different adaptations of ZIKV to hosts and vectors, and more importantly, this explains why NS1 K265-type strains have become predominantly endemic in nature. This study highlights the potential application in antiviral drug and vaccine development by targeting viral proteins' PTMs.
Collapse
Affiliation(s)
- Chenxiao Huang
- Institutes of Biology and Medical SciencesMOE Key Laboratory of Geriatric Diseases and ImmunologyJiangsu Key Laboratory of Infection and ImmunitySoochow UniversitySuzhou215000China
- Department of Clinical LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu School of Nanjing Medical UniversitySuzhou215000China
| | - Tao Jiang
- Institutes of Biology and Medical SciencesMOE Key Laboratory of Geriatric Diseases and ImmunologyJiangsu Key Laboratory of Infection and ImmunitySoochow UniversitySuzhou215000China
| | - Wen Pan
- Institutes of Biology and Medical SciencesMOE Key Laboratory of Geriatric Diseases and ImmunologyJiangsu Key Laboratory of Infection and ImmunitySoochow UniversitySuzhou215000China
| | - Tingting Feng
- Institutes of Biology and Medical SciencesMOE Key Laboratory of Geriatric Diseases and ImmunologyJiangsu Key Laboratory of Infection and ImmunitySoochow UniversitySuzhou215000China
| | - Xia Zhou
- School of Biology and Basic Medical ScienceSuzhou Medical College of Soochow UniversitySuzhou215000China
| | - Qihan Wu
- Shanghai‐MOST Key Laboratory of Health and Disease GenomicsNHC Key Lab of Reproduction RegulationShanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghai200000China
| | - Feng Ma
- National Key Laboratory of Immunity and Inflammationand CAMS Key Laboratory of Synthetic Biology Regulatory ElementsSuzhou Institute of Systems MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhou215123China
| | - Jianfeng Dai
- Institutes of Biology and Medical SciencesMOE Key Laboratory of Geriatric Diseases and ImmunologyJiangsu Key Laboratory of Infection and ImmunitySoochow UniversitySuzhou215000China
| |
Collapse
|
5
|
You S, Xu J, Guo Y, Guo X, Zhang Y, Zhang N, Sun G, Sun Y. E3 ubiquitin ligase WWP2 as a promising therapeutic target for diverse human diseases. Mol Aspects Med 2024; 96:101257. [PMID: 38430667 DOI: 10.1016/j.mam.2024.101257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
Mammalian E3 ubiquitin ligases have emerged in recent years as critical regulators of cellular homeostasis due to their roles in targeting substrate proteins for ubiquitination and triggering subsequent downstream signals. In this review, we describe the multiple roles of WWP2, an E3 ubiquitin ligase with unique and important functions in regulating a wide range of biological processes, including DNA repair, gene expression, signal transduction, and cell-fate decisions. As such, WWP2 has evolved to play a key role in normal physiology and diseases, such as tumorigenesis, skeletal development and diseases, immune regulation, cardiovascular disease, and others. We attempt to provide an overview of the biochemical, physiological, and pathophysiological roles of WWP2, as well as open questions for future research, particularly in the context of putative therapeutic opportunities.
Collapse
Affiliation(s)
- Shilong You
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiaqi Xu
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yushan Guo
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaofan Guo
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Zhang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China.
| | - Naijin Zhang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility, National Health Commission, China Medical University, Shenyang, Liaoning, China.
| | - Guozhe Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yingxian Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Yin Z, You S, Zhang S, Zhang L, Wu B, Huang X, Lu S, Cao L, Zhang Y, Li D, Zhang X, Liu J, Sun Y, Zhang N. Atorvastatin rescues vascular endothelial injury in hypertension by WWP2-mediated ubiquitination and degradation of ATP5A. Biomed Pharmacother 2023; 166:115228. [PMID: 37557013 DOI: 10.1016/j.biopha.2023.115228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
As a widely used lipid-lowering drug in clinical practice, atorvastatin is widely recognized for its role in protecting vascular endothelium in the cardiovascular system. However, a clear mechanistic understanding of its action is lacking. Here, we found that atorvastatin counteracted angiotensin II-induced vascular endothelial injury in mice with hypertension. Mechanistically, atorvastatin up-regulated WWP2, a E6AP C-terminus (HECT)-type E3 ubiquitin ligase with an essential role in regulating protein ubiquitination and various biological processes, thereby rescuing vascular endothelial injury. By ubiquitinating ATP5A (ATP synthase mitochondrial F1 complex subunit alpha), WWP2 degraded ATP5A via the proteasome pathway, stabilizing Bcl-2/Bax in the mitochondrial pathway of apoptosis. Moreover, atorvastatin further ameliorated death of vascular endothelial cells and improved vascular endothelial functions under WWP2 overexpression, whereas WWP2 knockout abrogated these beneficial effects of atorvastatin. Furthermore, we generated endothelial cell-specific WWP2 knockout mice, and this WWP2-mediated mechanism was faithfully recapitulated in vivo. Thus, we propose that activation of a WWP2-dependent pathway that is pathologically repressed in damaged vascular endothelium under hypertension is a major mechanism of atorvastatin. Our findings are also pertinent to develop novel therapeutic strategies for vascular endothelial injury-related cardiovascular diseases.
Collapse
Affiliation(s)
- Zeyu Yin
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shu Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Linlin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinyue Huang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Da Li
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, Liaoning Province, China; Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xingang Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jingwei Liu
- Department of Anus and Intestine Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, Liaoning Province, China.
| |
Collapse
|
7
|
You S, Xu J, Yin Z, Wu B, Wang P, Hao M, Cheng C, Liu M, Zhao Y, Jia P, Jiang H, Li D, Cao L, Zhang X, Zhang Y, Sun Y, Zhang N. Down-regulation of WWP2 aggravates Type 2 diabetes mellitus-induced vascular endothelial injury through modulating ubiquitination and degradation of DDX3X. Cardiovasc Diabetol 2023; 22:107. [PMID: 37149668 PMCID: PMC10164326 DOI: 10.1186/s12933-023-01818-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/29/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Endothelial injury caused by Type 2 diabetes mellitus (T2DM) is considered as a mainstay in the pathophysiology of diabetic vascular complications (DVCs). However, the molecular mechanism of T2DM-induced endothelial injury remains largely unknown. Here, we found that endothelial WW domain-containing E3 ubiquitin protein ligase 2 (WWP2) act as a novel regulator for T2DM-induced vascular endothelial injury through modulating ubiquitination and degradation of DEAD-box helicase 3 X-linked (DDX3X). METHODS Single-cell transcriptome analysis was used to evaluate WWP2 expression in vascular endothelial cells of T2DM patients and healthy controls. Endothelial-specific Wwp2 knockout mice were used to investigate the effect of WWP2 on T2DM-induced vascular endothelial injury. In vitro loss- and gain-of-function studies were performed to assess the function of WWP2 on cell proliferation and apoptosis of human umbilical vein endothelial cells. The substrate protein of WWP2 was verified using mass spectrometry, coimmunoprecipitation assays and immunofluorescence assays. The mechanism of WWP2 regulation on substrate protein was investigated by pulse-chase assay and ubiquitination assay. RESULTS The expression of WWP2 was significantly down-regulated in vascular endothelial cells during T2DM. Endothelial-specific Wwp2 knockout in mice significantly aggravated T2DM-induced vascular endothelial injury and vascular remodeling after endothelial injury. Our in vitro experiments showed that WWP2 protected against endothelial injury by promoting cell proliferation and inhibiting apoptosis in ECs. Mechanically, we found that WWP2 is down-regulated in high glucose and palmitic acid (HG/PA)-induced ECs due to c-Jun N-terminal kinase (JNK) activation, and uncovered that WWP2 suppresses HG/PA-induced endothelial injury by catalyzing K63-linked polyubiquitination of DDX3X and targeting it for proteasomal degradation. CONCLUSION Our studies revealed the key role of endothelial WWP2 and the fundamental importance of the JNK-WWP2-DDX3X regulatory axis in T2DM-induced vascular endothelial injury, suggesting that WWP2 may serve as a new therapeutic target for DVCs.
Collapse
Affiliation(s)
- Shilong You
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Jiaqi Xu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Zeyu Yin
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Boquan Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Pengbo Wang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Mingjun Hao
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004 China
| | - Cheng Cheng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Mengke Liu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Yuanhui Zhao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Hongkun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001 China
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004 China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, 110004 China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, 77 Puhe Road, Shenbei New District, Shenyang, 110001 Liaoning Province People’s Republic of China
- Institute of School of Basic Medicine, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Xingang Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
- Institute of School of Basic Medicine, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001 Liaoning Province People’s Republic of China
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province People’s Republic of China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, 110004 China
| |
Collapse
|
8
|
Zuo Q, Gong W, Yao Z, Xia Q, Zhang Y, Li B. Identification of key events and regulatory networks in the formation process of primordial germ cell based on proteomics. J Cell Physiol 2023; 238:610-630. [PMID: 36745473 DOI: 10.1002/jcp.30952] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Accepted: 01/09/2023] [Indexed: 02/07/2023]
Abstract
Currently, studies have analyzed the formation mechanism of primordial germ cell (PGC) at the transcriptional level, but few at the protein level, which made the mechanism study of PGC formation not systematic. Here, we screened differential expression proteins (DEPs) regulated PGC formation by label-free proteomics with a novel sampling strategy of embryonic stem cells and PGC. Analysis of DEPs showed that multiple key events were involved, such as the transition from glycolysis to oxidative phosphorylation, activation of autophagy, low DNA methylation ensured the normal formation of PGC, beyond that, protein ubiquitination also played an important role in PGC formation. Importantly, the progression of such events was attributed to the inconsistency between transcription and translation. Interestingly, MAPK, PPAR, Wnt, and JAK signaling pathways not only interact with each other but also interact with different events to participate in the formation of PGC, which formed the PGC regulatory network. According to the regulatory network, the efficiency of PGC formation in induction system can be significantly improved. In conclusion, our results indicate that chicken PGC formation is a complex process involving multiple events and signals, which provide technical support for the specific application in PGC research.
Collapse
Affiliation(s)
- Qisheng Zuo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Wei Gong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Zeling Yao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Qian Xia
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Yani Zhang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Bichun Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| |
Collapse
|
9
|
Targeting CSC-related transcription factors by E3 ubiquitin ligases for cancer therapy. Semin Cancer Biol 2022; 87:84-97. [PMID: 36371028 DOI: 10.1016/j.semcancer.2022.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/07/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Evidence has revealed that transcription factors play essential roles in regulation of multiple cellular processes, including cell proliferation, metastasis, EMT, cancer stem cells and chemoresistance. Dysregulated expression levels of transcription factors contribute to tumorigenesis and malignant progression. The expression of transcription factors is tightly governed by several signaling pathways, noncoding RNAs and E3 ubiquitin ligases. Cancer stem cells (CSCs) have been validated in regulation of tumor metastasis, reoccurrence and chemoresistance in human cancer. Transcription factors have been verified to participate in regulation of CSC formation, including Oct4, SOX2, KLF4, c-Myc, Nanog, GATA, SALL4, Bmi-1, OLIG2, POU3F2 and FOX proteins. In this review article, we will describe the critical role of CSC-related transcription factors. We will further discuss which E3 ligases regulate the degradation of these CSC-related transcription factors and their underlying mechanisms. We also mentioned the functions and mechanisms of EMT-associated transcription factors such as ZEB1, ZEB2, Snail, Slug, Twist1 and Twist2. Furthermore, we highlight the therapeutic potential via targeting E3 ubiquitin ligases for modulation of these transcription factors.
Collapse
|
10
|
Li Q, Zhang W. Progress in Anticancer Drug Development Targeting Ubiquitination-Related Factors. Int J Mol Sci 2022; 23:ijms232315104. [PMID: 36499442 PMCID: PMC9737479 DOI: 10.3390/ijms232315104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 12/05/2022] Open
Abstract
Ubiquitination is extensively involved in critical signaling pathways through monitoring protein stability, subcellular localization, and activity. Dysregulation of this process results in severe diseases including malignant cancers. To develop drugs targeting ubiquitination-related factors is a hotspot in research to realize better therapy of human diseases. Ubiquitination comprises three successive reactions mediated by Ub-activating enzyme E1, Ub-conjugating enzyme E2, and Ub ligase E3. As expected, multiple ubiquitination enzymes have been highlighted as targets for anticancer drug development due to their dominant effect on tumorigenesis and cancer progression. In this review, we discuss recent progresses in anticancer drug development targeting enzymatic machinery components.
Collapse
|
11
|
Lu X, Xu H, Xu J, Lu S, You S, Huang X, Zhang N, Zhang L. The regulatory roles of the E3 ubiquitin ligase NEDD4 family in DNA damage response. Front Physiol 2022; 13:968927. [PMID: 36091384 PMCID: PMC9458852 DOI: 10.3389/fphys.2022.968927] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
E3 ubiquitin ligases, an important part of ubiquitin proteasome system, catalyze the covalent binding of ubiquitin to target substrates, which plays a role in protein ubiquitination and regulates different biological process. DNA damage response (DDR) is induced in response to DNA damage to maintain genome integrity and stability, and this process has crucial significance to a series of cell activities such as differentiation, apoptosis, cell cycle. The NEDD4 family, belonging to HECT E3 ubiquitin ligases, is reported as regulators that participate in the DDR process by recognizing different substrates. In this review, we summarize recent researches on NEDD4 family members in the DDR and discuss the roles of NEDD4 family members in the cascade reactions induced by DNA damage. This review may contribute to the further study of pathophysiology for certain diseases and pharmacology for targeted drugs.
Collapse
Affiliation(s)
- Xinxin Lu
- Department of Hematology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Haiqi Xu
- Department of Hematology, General Hospital of PLA Northern Theater Command, Shenyang, LN, China
| | - Jiaqi Xu
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Saien Lu
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Shilong You
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Xinyue Huang
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Naijin Zhang
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Lijun Zhang
- Department of Hematology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| |
Collapse
|
12
|
Wang X, Ma L, Zhang S, Song Q, He X, Wang J. WWP2 ameliorates oxidative stress and inflammation in atherosclerotic mice through regulation of PDCD4/HO-1 pathway. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1057-1067. [PMID: 35983977 PMCID: PMC9828489 DOI: 10.3724/abbs.2022091] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
WWP2 is a HECT-type E3 ubiquitin ligase that regulates various physiological and pathological activities by binding to different substrates, but its role in atherosclerosis (AS) remains largely unknown. The objective of the present study is to investigate the role and underlying molecular mechanisms of WWP2 in endothelial injury. We found that WWP2 expression is significantly decreased in Apolipoprotein E (ApoE) -/- mice. Overexpression of WWP2 attenuates oxidative stress and inflammation in AS mice, while knockdown of WWP2 has opposite effects. WWP2 overexpression alleviates oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cell (HUVEC) injury, evidenced by the decreased oxidative stress levels and the secretion of inflammatory cytokines. Programmed cell death 4 (PDCD4) is identified as a potential substrate of WWP2. Co-immunoprecipitation (Co-IP) further demonstrates that WWP2 interacts with PDCD4, which is enhanced by ox-LDL treatment. Furthermore, the level of PDCD4 ubiquitination is significantly increased by WWP2 overexpression under the condition of MG132 treatment, while WWP2 knockdown shows opposite results. Subsequently, rescue experiments demonstrate that WWP2 knockdown further aggravates oxidative stress and inflammation in ox-LDL-treated HUVECs, while knockdown of PDCD4 alleviates this effect. Moreover, the use of sn-protoporphyrin (SnPP), an inhibitor of HO-1 pathway, confirms that PDCD4 enhances endothelial injury induced by ox-LDL through inhibiting HO-1 pathway. In conclusion, our results suggest that WWP2 protects against atherosclerosis progression via the PDCD4/HO-1 pathway, which may provide a novel treatment strategy for atherosclerosis.
Collapse
Affiliation(s)
- Xingye Wang
- Department of Structural Cardiologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Lu Ma
- Department of Graduate SchoolXi’an Shiyou UniversityXi’an710065China
| | - Songlin Zhang
- Department of Structural Cardiologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Qiang Song
- Department of Structural Cardiologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Xumei He
- Department of Structural Cardiologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Jun Wang
- Department of Structural Cardiologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China,Correspondence address. Tel: +86-29-85434128; E-mail:
| |
Collapse
|
13
|
Mamun MMA, Khan MR, Zhu Y, Zhang Y, Zhou S, Xu R, Bukhari I, Thorne RF, Li J, Zhang XD, Liu G, Chen S, Wu M, Song X. Stub1 maintains proteostasis of master transcription factors in embryonic stem cells. Cell Rep 2022; 39:110919. [PMID: 35675767 DOI: 10.1016/j.celrep.2022.110919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/01/2022] [Accepted: 05/10/2022] [Indexed: 12/01/2022] Open
Abstract
The pluripotency and differentiation states of embryonic stem cells (ESCs) are regulated by a set of core transcription factors, primarily Sox2, Oct4, and Nanog. Although their transcriptional regulation has been studied extensively, the contribution of posttranslational modifications in Sox2, Oct4, and Nanog are poorly understood. Here, using a CRISPR-Cas9 knockout library screen in murine ESCs, we identify the E3 ubiquitin ligase Stub1 as a negative regulator of pluripotency. Manipulation of Stub1 expression in murine ESCs shows that ectopic Stub1 expression significantly reduces the protein half-life of Sox2, Oct4, and Nanog. Mechanistic investigations reveal Stub1 catalyzes the polyubiquitination and 26S proteasomal degradation of Sox2 and Nanog through K48-linked ubiquitin chains and Oct4 via K63 linkage. Stub1 deficiency positively enhances somatic cell reprogramming and delays differentiation, whereas its enforced expression triggers ESC differentiation. The discovery of Stub1 as an integral pluripotency regulator strengthens our understanding of ESC regulation beyond conventional transcriptional control mechanisms.
Collapse
Affiliation(s)
- Md Mahfuz Al Mamun
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Muhammad Riaz Khan
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8 Canada
| | - Yifu Zhu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China
| | - Yuwei Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China
| | - Shuai Zhou
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Ran Xu
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Ihtisham Bukhari
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China; School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2258, Australia
| | - Jinming Li
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China; School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Guangzhi Liu
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China.
| | - Song Chen
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China; Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, Jiangsu 223300, China.
| | - Mian Wu
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China.
| | - Xiaoyuan Song
- MOE Key Laboratory for Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
14
|
Bakhmet EI, Tomilin AN. Key features of the POU transcription factor Oct4 from an evolutionary perspective. Cell Mol Life Sci 2021; 78:7339-7353. [PMID: 34698883 PMCID: PMC11072838 DOI: 10.1007/s00018-021-03975-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/16/2021] [Accepted: 10/12/2021] [Indexed: 01/06/2023]
Abstract
Oct4, a class V POU-domain protein that is encoded by the Pou5f1 gene, is thought to be a key transcription factor in the early development of mammals. This transcription factor plays indispensable roles in pluripotent stem cells as well as in the acquisition of pluripotency during somatic cell reprogramming. Oct4 has also been shown to play a role as a pioneer transcription factor during zygotic genome activation (ZGA) from zebrafish to human. However, during the past decade, several studies have brought these conclusions into question. It was clearly shown that the first steps in mouse development are not affected by the loss of Oct4. Subsequently, the role of Oct4 as a genome activator was brought into doubt. It was also found that the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) could proceed without Oct4. In this review, we summarize recent findings, reassess the role of Oct4 in reprogramming and ZGA, and point to structural features that may underlie this role. We speculate that pluripotent stem cells resemble neural stem cells more closely than previously thought. Oct4 orthologs within the POUV class hold key roles in genome activation during early development of species with late ZGA. However, in Placentalia, eutherian-specific proteins such as Dux overtake Oct4 in ZGA and endow them with the formation of an evolutionary new tissue-the placenta.
Collapse
Affiliation(s)
- Evgeny I Bakhmet
- Laboratory of the Molecular Biology of Stem Cells, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia.
| | - Alexey N Tomilin
- Laboratory of the Molecular Biology of Stem Cells, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
15
|
Ye P, Chi X, Cha JH, Luo S, Yang G, Yan X, Yang WH. Potential of E3 Ubiquitin Ligases in Cancer Immunity: Opportunities and Challenges. Cells 2021; 10:cells10123309. [PMID: 34943817 PMCID: PMC8699390 DOI: 10.3390/cells10123309] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer immunotherapies, including immune checkpoint inhibitors and immune pathway–targeted therapies, are promising clinical strategies for treating cancer. However, drug resistance and adverse reactions remain the main challenges for immunotherapy management. The future direction of immunotherapy is mainly to reduce side effects and improve the treatment response rate by finding new targets and new methods of combination therapy. Ubiquitination plays a crucial role in regulating the degradation of immune checkpoints and the activation of immune-related pathways. Some drugs that target E3 ubiquitin ligases have exhibited beneficial effects in preclinical and clinical antitumor treatments. In this review, we discuss mechanisms through which E3 ligases regulate tumor immune checkpoints and immune-related pathways as well as the opportunities and challenges for integrating E3 ligases targeting drugs into cancer immunotherapy.
Collapse
Affiliation(s)
- Peng Ye
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Xiaoxia Chi
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Jong-Ho Cha
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Korea;
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea
| | - Shahang Luo
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Guanghui Yang
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
| | - Xiuwen Yan
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
- Correspondence: (X.Y.); (W.-H.Y.)
| | - Wen-Hao Yang
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes and Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou 910095, China; (P.Y.); (X.C.); (S.L.); (G.Y.)
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Correspondence: (X.Y.); (W.-H.Y.)
| |
Collapse
|
16
|
Bou G, Guo J, Fang Y, Li X, Wei R, Li Y, Liu Z. Interspecies cell fusion between mouse embryonic stem cell and porcine pluripotent cell. Reprod Domest Anim 2021; 56:1095-1103. [PMID: 33993554 DOI: 10.1111/rda.13952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/12/2021] [Indexed: 11/30/2022]
Abstract
In the area of stem cell research, fusion of somatic cells into pluripotent cells such as mouse embryonic stem (ES) cells induces reprogramming of the somatic nucleus and can be used to study the effect of trans-acting factors from the pluripotent cell on the pluripotent state of somatic nucleus. As many other groups, we previously established a porcine pluripotent cell line at a low potential. Therefore, here, we performed experiments to investigate if the fusion with mouse ES cell could improve the pluripotent state of porcine pluripotent cell. Our data showed that resultant mouse-porcine interspecies fused cells are AP positive, and could be passaged up to 20 passages. Different degrees of increases in expression of porcine pluripotent genes proved that pig-origin gene network can be programmed by mouse ES. Further differentiation study also confirmed these fused cells' potential to form three germ layers. However, unexpectedly, we found that chromosome loss and aberrant (especially in porcine chromosomes) is severe after the cell fusion, implying that interspecies cell fusion may be not suitable to study porcine pluripotency without additional supportive conditions for genome stabilization.
Collapse
Affiliation(s)
- Gerelchimeg Bou
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Jia Guo
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yuan Fang
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xuechun Li
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Renyue Wei
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yan Li
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
17
|
Ubiquitin-Specific Protease 3 Deubiquitinates and Stabilizes Oct4 Protein in Human Embryonic Stem Cells. Int J Mol Sci 2021; 22:ijms22115584. [PMID: 34070420 PMCID: PMC8197518 DOI: 10.3390/ijms22115584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 11/16/2022] Open
Abstract
Oct4 is an important mammalian POU family transcription factor expressed by early human embryonic stem cells (hESCs). The precise level of Oct4 governs the pluripotency and fate determination of hESCs. Several post-translational modifications (PTMs) of Oct4 including phosphorylation, ubiquitination, and SUMOylation have been reported to regulate its critical functions in hESCs. Ubiquitination and deubiquitination of Oct4 should be well balanced to maintain the pluripotency of hESCs. The protein turnover of Oct4 is regulated by several E3 ligases through ubiquitin-mediated degradation. However, reversal of ubiquitination by deubiquitinating enzymes (DUBs) has not been reported for Oct4. In this study, we generated a ubiquitin-specific protease 3 (USP3) gene knockout using the CRISPR/Cas9 system and demonstrated that USP3 acts as a protein stabilizer of Oct4 by deubiquitinating Oct4. USP3 interacts with endogenous Oct4 and co-localizes in the nucleus of hESCs. The depletion of USP3 leads to a decrease in Oct4 protein level and loss of pluripotent morphology in hESCs. Thus, our results show that USP3 plays an important role in controlling optimum protein level of Oct4 to retain pluripotency of hESCs.
Collapse
|
18
|
Wu Y, Zhang W. The Role of E3s in Regulating Pluripotency of Embryonic Stem Cells and Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:1168. [PMID: 33503896 PMCID: PMC7865285 DOI: 10.3390/ijms22031168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
Pluripotent embryonic stem cells (ESCs) are derived from early embryos and can differentiate into any type of cells in living organisms. Induced pluripotent stem cells (iPSCs) resemble ESCs, both of which serve as excellent sources to study early embryonic development and realize cell replacement therapies for age-related degenerative diseases and other cell dysfunction-related illnesses. To achieve these valuable applications, comprehensively understanding of the mechanisms underlying pluripotency maintenance and acquisition is critical. Ubiquitination modifies proteins with Ubiquitin (Ub) at the post-translational level to monitor protein stability and activity. It is extensively involved in pluripotency-specific regulatory networks in ESCs and iPSCs. Ubiquitination is achieved by sequential actions of the Ub-activating enzyme E1, Ub-conjugating enzyme E2, and Ub ligase E3. Compared with E1s and E2s, E3s are most abundant, responsible for substrate selectivity and functional diversity. In this review, we focus on E3 ligases to discuss recent progresses in understanding how they regulate pluripotency and somatic cell reprogramming through ubiquitinating core ESC regulators.
Collapse
Affiliation(s)
| | - Weiwei Zhang
- College of Life Sciences, Capital Normal University, Beijing 100048, China;
| |
Collapse
|
19
|
Fu J, Zheng H, Xue Y, Jin R, Yang G, Chen Z, Yuan G. WWP2 Promotes Odontoblastic Differentiation by Monoubiquitinating KLF5. J Dent Res 2020; 100:432-439. [PMID: 33164644 DOI: 10.1177/0022034520970866] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
WW domain-containing E3 Ub-protein ligase 2 (WWP2) belongs to the homologous to E6AP C-terminus (HECT) E3 ligase family. It has been explored to regulate osteogenic differentiation, chondrogenesis, and palatogenesis. Odontoblasts are terminally differentiated mesenchymal cells, which contribute to dentin formation in tooth development. However, it remained unknown whether WWP2 participated in odontoblast differentiation. In this study, WWP2 was found to be expressed in mouse dental papilla cells (mDPCs), odontoblasts, and odontoblastic-induced mDPCs by immunohistochemistry and Western blotting. Besides, WWP2 expression was decreased in the cytoplasm but increased in the nuclei of differentiation-induced mDPCs. When Wwp2 was knocked down, the elevated expression of odontoblast marker genes (Dmp1 and Dspp) in mDPCs induced by differentiation medium was suppressed. Meanwhile, a decrease of alkaline phosphatase (ALP) activity was observed by ALP staining, and reduced formation of mineralized matrix nodules was demonstrated by Alizarin Red S staining. Overexpression of WWP2 presented opposite results to knockdown experiments, suggesting that WWP2 promoted odontoblastic differentiation of mDPCs. Further investigation found that WWP2 was coexpressed and interacted with KLF5 in the nuclei, leading to ubiquitination of KLF5. The PPPSY (PY2) motif of KLF5 was essential for its physical binding with WWP2. Also, cysteine 838 (Cys838) of WWP2 was the active site for ubiquitination of KLF5, which did not lead to proteolysis of KLF5. Then, KLF5 was confirmed to be monoubiquitinated and transactivated by WWP2, which promoted the expression of KLF5 downstream genes Dmp1 and Dspp. Deletion of the PY2 motif of KLF5 or mutation of Cys838 of WWP2 reduced the upregulation of Dmp1 and Dspp. Besides, lysine (K) residues K31, K52, K83, and K265 of KLF5 were verified to be crucial to WWP2-mediated KLF5 transactivation. Taken together, WWP2 promoted odontoblastic differentiation by monoubiquitinating KLF5.
Collapse
Affiliation(s)
- J Fu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, HuBei, China
| | - H Zheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, HuBei, China
| | - Y Xue
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, HuBei, China
| | - R Jin
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, HuBei, China
| | - G Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, HuBei, China
| | - Z Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, HuBei, China
| | - G Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, HuBei, China
| |
Collapse
|
20
|
Kaushal K, Ramakrishna S. Deubiquitinating Enzyme-Mediated Signaling Networks in Cancer Stem Cells. Cancers (Basel) 2020; 12:E3253. [PMID: 33158118 PMCID: PMC7694198 DOI: 10.3390/cancers12113253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) have both the capacity for self-renewal and the potential to differentiate and contribute to multiple tumor properties, such as recurrence, metastasis, heterogeneity, multidrug resistance, and radiation resistance. Thus, CSCs are considered to be promising therapeutic targets for cancer therapy. The function of CSCs can be regulated by ubiquitination and deubiquitination of proteins related to the specific stemness of the cells executing various stem cell fate choices. To regulate the balance between ubiquitination and deubiquitination processes, the disassembly of ubiquitin chains from specific substrates by deubiquitinating enzymes (DUBs) is crucial. Several key developmental and signaling pathways have been shown to play essential roles in this regulation. Growing evidence suggests that overactive or abnormal signaling within and among these pathways may contribute to the survival of CSCs. These signaling pathways have been experimentally shown to mediate various stem cell properties, such as self-renewal, cell fate decisions, survival, proliferation, and differentiation. In this review, we focus on the DUBs involved in CSCs signaling pathways, which are vital in regulating their stem-cell fate determination.
Collapse
Affiliation(s)
- Kamini Kaushal
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea;
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea;
- College of Medicine, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
21
|
Liu L, Zhang R, Wang X, Zhu H, Tian Z. Transcriptome analysis reveals molecular mechanisms responsive to acute cold stress in the tropical stenothermal fish tiger barb (Puntius tetrazona). BMC Genomics 2020; 21:737. [PMID: 33096997 PMCID: PMC7584086 DOI: 10.1186/s12864-020-07139-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 10/11/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Tropical stenothermal fish exhibit special tolerance and response to cold stress. However current knowledge of the molecular mechanisms response to cold stress in aquatic ectotherms is largely drawn from eurythermal or extreme stenothermal species. The tiger barb Puntius tetrazona is a tropical stenothermal fish, with great popularity in aquarium trade and research. RESULTS To investigate the response mechanism of P. tetrazona to low temperature, fish were exposed to increasing levels of acute cold stress. Histopathological analysis showed that the brain, gill, liver and muscle tissues appeared serious damage after cold stress (13 °C). Brain, gill, liver and muscle tissues from control (CTRL) groups (27 °C) and COLD stress groups (13 °C) of eight-month fish (gender-neutral) were sampled and assessed for transcriptomic profiling by high-throughput sequencing. 83.0 Gb of raw data were generated, filtered and assembled for de novo transcriptome assembly. According to the transcriptome reference, we obtained 392,878 transcripts and 238,878 unigenes, of which 89.29% of the latter were annotated. There were 23,743 differently expressed genes (DEGs) been filtered from four pairs of tissues (brain, gill, liver and muscle) between these cold stress and control groups. These DEGs were mainly involved in circadian entrainment, circadian rhythm, biosynthesis of steroid and fatty acid. There were 64 shared DEGs between the four pairs of groups, and five were related to ubiquitylation/deubiquitylation. Our results suggested that ubiquitin-mediated protein degradation might be necessary for tropical stenothermal fish coping with acute cold stress. Also, the significant cold-induced expression of heat shock 70 kDa protein (HSP70) and cold-induced RNA-binding protein (CIRBP) was verified. These results suggested that the expression of the molecular chaperones HSP70 and CIRBP in P. tetrazona might play a critical role in coping with acute cold stress. CONCLUSIONS This is the first transcriptome analysis of P. tetrazona using RNA-Seq technology. Novel findings about tropical stenothermal fish under cold stress (such as HSP70 and CIRBP genes) are presented here. This study contributes new insights into the molecular mechanisms of tropical stenothermal species response to acute cold stress.
Collapse
Affiliation(s)
- Lili Liu
- Beijing Key Laboratory of Fishery Biotechnology, Beijing Fisheries Research Institute, Beijing, 100068 China
| | - Rong Zhang
- Beijing Key Laboratory of Fishery Biotechnology, Beijing Fisheries Research Institute, Beijing, 100068 China
| | - Xiaowen Wang
- Beijing Key Laboratory of Fishery Biotechnology, Beijing Fisheries Research Institute, Beijing, 100068 China
| | - Hua Zhu
- Beijing Key Laboratory of Fishery Biotechnology, Beijing Fisheries Research Institute, Beijing, 100068 China
| | - Zhaohui Tian
- Beijing Key Laboratory of Fishery Biotechnology, Beijing Fisheries Research Institute, Beijing, 100068 China
| |
Collapse
|
22
|
Abstract
Derivation of induced Pluripotent Stem Cells (iPSCs) by reprogramming somatic cells to a pluripotent state has revolutionized stem cell research. Ensuing this, various groups have used genetic and non-genetic approaches to generate iPSCs from numerous cell types. However, achieving a pluripotent state in most of the reprogramming studies is marred by serious limitations such as low reprogramming efficiency and slow kinetics. These limitations are mainly due to the presence of potent barriers that exist during reprogramming when a mature cell is coaxed to achieve a pluripotent state. Several studies have revealed that intrinsic factors such as non-optimal stoichiometry of reprogramming factors, specific signaling pathways, cellular senescence, pluripotency-inhibiting transcription factors and microRNAs act as a roadblock. In addition, the epigenetic state of somatic cells and specific epigenetic modifications that occur during reprogramming also remarkably impede the generation of iPSCs. In this review, we present a comprehensive overview of the barriers that inhibit reprogramming and the understanding of which will pave the way to develop safe strategies for efficient reprogramming.
Collapse
|
23
|
Zhang Y, Qian H, Wu B, You S, Wu S, Lu S, Wang P, Cao L, Zhang N, Sun Y. E3 Ubiquitin ligase NEDD4 family‑regulatory network in cardiovascular disease. Int J Biol Sci 2020; 16:2727-2740. [PMID: 33110392 PMCID: PMC7586430 DOI: 10.7150/ijbs.48437] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
Protein ubiquitination represents a critical modification occurring after translation. E3 ligase catalyzes the covalent binding of ubiquitin to the protein substrate, which could be degraded. Ubiquitination as an important protein post-translational modification is closely related to cardiovascular disease. The NEDD4 family, belonging to HECT class of E3 ubiquitin ligases can recognize different substrate proteins, including PTEN, ENaC, Nav1.5, SMAD2, PARP1, Septin4, ALK1, SERCA2a, TGFβR3 and so on, via the WW domain to catalyze ubiquitination, thus participating in multiple cardiovascular-related disease such as hypertension, arrhythmia, myocardial infarction, heart failure, cardiotoxicity, cardiac hypertrophy, myocardial fibrosis, cardiac remodeling, atherosclerosis, pulmonary hypertension and heart valve disease. However, there is currently no review comprehensively clarifying the important role of NEDD4 family proteins in the cardiovascular system. Therefore, the present review summarized recent studies about NEDD4 family members in cardiovascular disease, providing novel insights into the prevention and treatment of cardiovascular disease. In addition, assessing transgenic animals and performing gene silencing would further identify the ubiquitination targets of NEDD4. NEDD4 quantification in clinical samples would also constitute an important method for determining NEDD4 significance in cardiovascular disease.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Qian
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shaojun Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Pingyuan Wang
- Staff scientist, Center for Molecular Medicine National Heart Lung and Blood Institute, National Institutes of Health, the United States
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
24
|
Alix E, Godlee C, Cerny O, Blundell S, Tocci R, Matthews S, Liu M, Pruneda JN, Swatek KN, Komander D, Sleap T, Holden DW. The Tumour Suppressor TMEM127 Is a Nedd4-Family E3 Ligase Adaptor Required by Salmonella SteD to Ubiquitinate and Degrade MHC Class II Molecules. Cell Host Microbe 2020; 28:54-68.e7. [PMID: 32526160 PMCID: PMC7342019 DOI: 10.1016/j.chom.2020.04.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/13/2020] [Accepted: 04/29/2020] [Indexed: 12/21/2022]
Abstract
The Salmonella enterica effector SteD depletes mature MHC class II (mMHCII) molecules from the surface of infected antigen-presenting cells through ubiquitination of the cytoplasmic tail of the mMHCII β chain. Here, through a genome-wide mutant screen of human antigen-presenting cells, we show that the NEDD4 family HECT E3 ubiquitin ligase WWP2 and a tumor-suppressing transmembrane protein of unknown biochemical function, TMEM127, are required for SteD-dependent ubiquitination of mMHCII. Although evidently not involved in normal regulation of mMHCII, TMEM127 was essential for SteD to suppress both mMHCII antigen presentation in mouse dendritic cells and MHCII-dependent CD4+ T cell activation. We found that TMEM127 contains a canonical PPxY motif, which was required for binding to WWP2. SteD bound to TMEM127 and enabled TMEM127 to interact with and induce ubiquitination of mature MHCII. Furthermore, SteD also underwent TMEM127- and WWP2-dependent ubiquitination, which both contributed to its degradation and augmented its activity on mMHCII.
Collapse
Affiliation(s)
- Eric Alix
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - Camilla Godlee
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - Ondrej Cerny
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - Samkeliso Blundell
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - Romina Tocci
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - Sophie Matthews
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - Mei Liu
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - Jonathan N Pruneda
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Kirby N Swatek
- Ubiquitin Signalling Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royale Parade, 3052 Parkville, Melbourne, Australia
| | - David Komander
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Tabitha Sleap
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK
| | - David W Holden
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK.
| |
Collapse
|
25
|
Zhang Y, You S, Tian Y, Lu S, Cao L, Sun Y, Zhang N. WWP2 regulates SIRT1-STAT3 acetylation and phosphorylation involved in hypertensive angiopathy. J Cell Mol Med 2020; 24:9041-9054. [PMID: 32627301 PMCID: PMC7417706 DOI: 10.1111/jcmm.15538] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/23/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
WWP2 is a HECT‐type E3 ubiquitin ligase that regulates various physiological and pathological activities by binding to different substrates, but its function and regulatory mechanism in vascular smooth muscle cells (VSMCs) are still unknown. Here, we clarified the role of WWP2 in the regulation of SIRT1‐STAT3 and the impact of this regulatory process in VSMCs. We demonstrated that WWP2 expression was significantly increased in angiotensin II‐induced VSMCs model. Knockdown of WWP2 significantly inhibited angiotensin II‐induced VSMCs proliferation, migration and phenotypic transformation, whereas overexpression of WWP2 had opposite effects. In vivo experiments showed that vascular smooth muscle‐specific WWP2 knockout mice significantly relieved angiotensin II‐induced hypertensive angiopathy. Mechanistically, mass spectrometry and co‐immunoprecipitation assays identified that WWP2 is a novel interacting protein of SIRT1 and STAT3. Moreover, WWP2 formed a complex with SIRT1‐STAT3, inhibiting the interaction between SIRT1 and STAT3, then reducing the inhibitory effect of SIRT1 on STAT3, ensuing promoting STAT3‐K685 acetylation and STAT3‐Y705 phosphorylation in angiotensin II‐induced VSMCs and mice. In conclusion, WWP2 modulates hypertensive angiopathy by regulating SIRT1‐STAT3 and WWP2 suppression in VSMCs can alleviate hypertensive angiopathy vitro and vivo. These findings provide new insights into the treatment of hypertensive vascular diseases.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shilong You
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yichen Tian
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Saien Lu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, Institute of Translational Medicine, China Medical University, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Cellular Functions of OCT-3/4 Regulated by Ubiquitination in Proliferating Cells. Cancers (Basel) 2020; 12:cancers12030663. [PMID: 32178477 PMCID: PMC7139964 DOI: 10.3390/cancers12030663] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/18/2022] Open
Abstract
Octamer-binding transcription factor 3/4 (OCT-3/4), which is involved in the tumorigenesis of somatic cancers, has diverse functions during cancer development. Overexpression of OCT-3/4 has been detected in various human somatic tumors, indicating that OCT-3/4 activation may contribute to the development and progression of cancers. Stem cells can undergo self-renewal, pluripotency, and reprogramming with the help of at least four transcription factors, OCT-3/4, SRY box-containing gene 2 (SOX2), Krüppel-like factor 4 (KLF4), and c-MYC. Of these, OCT-3/4 plays a critical role in maintenance of undifferentiated state of embryonic stem cells (ESCs) and in production of induced pluripotent stem cells (iPSCs). Stem cells can undergo partitioning through mitosis and separate into specific cell types, three embryonic germ layers: the endoderm, the mesoderm, and the trophectoderm. It has been demonstrated that the stability of OCT-3/4 is mediated by the ubiquitin-proteasome system (UPS), which is one of the key cellular mechanisms for cellular homeostasis. The framework of the mechanism is simple, but the proteolytic machinery is complicated. Ubiquitination promotes protein degradation, and ubiquitination of OCT-3/4 leads to regulation of cellular proliferation and differentiation. Therefore, it is expected that OCT-3/4 may play a key role in proliferation and differentiation of proliferating cells.
Collapse
|
27
|
Selective targeting of ubiquitination and degradation of PARP1 by E3 ubiquitin ligase WWP2 regulates isoproterenol-induced cardiac remodeling. Cell Death Differ 2020; 27:2605-2619. [PMID: 32139900 DOI: 10.1038/s41418-020-0523-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
The elevated expression of poly(ADP-ribose) polymerase-1 (PARP1) and increased PARP1 activity, namely, poly(ADP-ribosyl)ation (PARylation), have been observed in cardiac remodeling, leading to extreme energy consumption and myocardial damage. However, the mechanisms underlying the regulation of PARP1 require further study. WWP2, a HECT-type E3 ubiquitin ligase, is highly expressed in the heart, but its function there is largely unknown. Here, we clarified the role of WWP2 in the regulation of PARP1 and the impact of this regulatory process on cardiac remodeling. We determined that the knockout of WWP2 specifically in myocardium decreased the level of PARP1 ubiquitination and increased the effects of isoproterenol (ISO)-induced PARP1 and PARylation, in turn aggravating ISO-induced myocardial hypertrophy, heart failure, and myocardial fibrosis. Similar findings were obtained in a model of ISO-induced H9c2 cells with WWP2 knockdown, while the reexpression of WWP2 significantly increased PARP1 ubiquitination and decreased PAPR1 and PARylation levels. Mechanistically, coimmunoprecipitation results identified that WWP2 is a novel interacting protein of PARP1 and mainly interacts with its BRCT domain, thus mediating the degradation of PARP1 through the ubiquitin-proteasome system. In addition, lysine 418 (K418) and lysine 249 (K249) were shown to be of critical importance in regulating PARP1 ubiquitination and degradation by WWP2. These findings reveal a novel WWP2-PARP1 signal transduction pathway involved in controlling cardiac remodeling and may provide a basis for exploring new strategies for treating heart disorders related to cardiac remodeling.
Collapse
|
28
|
de Dieuleveult M, Miotto B. Ubiquitin Dynamics in Stem Cell Biology: Current Challenges and Perspectives. Bioessays 2020; 42:e1900129. [PMID: 31967345 DOI: 10.1002/bies.201900129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/18/2019] [Indexed: 11/09/2022]
Abstract
Ubiquitination plays a central role in the regulation of stem cell self-renewal, propagation, and differentiation. In this review, the functions of ubiquitin dynamics in a myriad of cellular processes, acting along side the pluripotency network, to regulate embryonic stem cell identity are highlighted. The implication of deubiquitinases (DUBs) and E3 Ubiquitin (Ub) ligases in cellular functions beyond protein degradation is reported, including key functions in the regulation of mRNA stability, protein translation, and intra-cellular trafficking; and how it affects cell metabolism, the micro-environment, and chromatin organization is discussed. Finally, unsolved issues in the field are emphasized and will need to be tackled in order to fully understand the contribution of ubiquitin dynamics to stem cell self-renewal and differentiation.
Collapse
Affiliation(s)
- Maud de Dieuleveult
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014, Paris, France
| | - Benoit Miotto
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, F-75014, Paris, France
| |
Collapse
|
29
|
The Role of Ubiquitination in Regulating Embryonic Stem Cell Maintenance and Cancer Development. Int J Mol Sci 2019; 20:ijms20112667. [PMID: 31151253 PMCID: PMC6600158 DOI: 10.3390/ijms20112667] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/19/2019] [Accepted: 05/28/2019] [Indexed: 12/18/2022] Open
Abstract
Ubiquitination regulates nearly every aspect of cellular events in eukaryotes. It modifies intracellular proteins with 76-amino acid polypeptide ubiquitin (Ub) and destines them for proteolysis or activity alteration. Ubiquitination is generally achieved by a tri-enzyme machinery involving ubiquitin activating enzymes (E1), ubiquitin conjugating enzymes (E2) and ubiquitin ligases (E3). E1 activates Ub and transfers it to the active cysteine site of E2 via a transesterification reaction. E3 coordinates with E2 to mediate isopeptide bond formation between Ub and substrate protein. The E1-E2-E3 cascade can create diverse types of Ub modifications, hence effecting distinct outcomes on the substrate proteins. Dysregulation of ubiquitination results in severe consequences and human diseases. There include cancers, developmental defects and immune disorders. In this review, we provide an overview of the ubiquitination machinery and discuss the recent progresses in the ubiquitination-mediated regulation of embryonic stem cell maintenance and cancer biology.
Collapse
|
30
|
Zhang R, Zhang J, Luo W, Luo Z, Shi S. WWP2 Is One Promising Novel Oncogene. Pathol Oncol Res 2018; 25:443-446. [PMID: 30415470 DOI: 10.1007/s12253-018-0506-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 10/15/2018] [Indexed: 12/25/2022]
Abstract
WWP2 is an E3 ubiquitin ligase and plays an important role in regulation of many cellular biological activities through ubiquitination and degradation of its substrates. Recently accumulating evidences indicate that WWP2 plays a crucial part in the pathogenesis in different types of tumors. In this report, the role of this gene especially in tumorigenesis was reviewed. WWP2 is dysregulated in various of tumors, and it promotes carcinogenesis mainly through PTEN/Akt signaling pathway. WWP2 also participates in anti-cancer agents' sensitivity, indicating WWP2 may be a novel target for cancer treatment. WWP2 is one promising novel oncogene.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Thoracic Surgery, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan, 640021, People's Republic of China
| | - Jianwu Zhang
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, 637100, People's Republic of China
| | - Wei Luo
- Department of Respiratory Medicine, The People's Hospital of Leshan, Leshan, Sichuan, 640000, People's Republic of China
| | - Zhuang Luo
- Department of Pumnary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, People's Republic of China.
| | - Shaoqing Shi
- Department of Pumnary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, People's Republic of China.
| |
Collapse
|
31
|
Li S, Xiao F, Zhang J, Sun X, Wang H, Zeng Y, Hu J, Tang F, Gu J, Zhao Y, Jin Y, Liao B. Disruption of OCT4 Ubiquitination Increases OCT4 Protein Stability and ASH2L-B-Mediated H3K4 Methylation Promoting Pluripotency Acquisition. Stem Cell Reports 2018; 11:973-987. [PMID: 30269953 PMCID: PMC6178847 DOI: 10.1016/j.stemcr.2018.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 11/25/2022] Open
Abstract
The protein level of OCT4, a core pluripotency transcription factor, is vital for embryonic stem cell (ESC) maintenance, differentiation, and somatic cell reprogramming. However, how OCT4 protein levels are controlled during reprogramming remains largely unknown. Here, we identify ubiquitin conjugation sites of OCT4 and report that disruption of WWP2-catalyzed OCT4 ubiquitination or ablation of Wwp2 significantly promotes the efficiency of pluripotency induction from mouse embryonic fibroblasts. Mechanistically, disruption of WWP2-mediated OCT4 ubiquitination elevates OCT4 protein stability and H3K4 methylation level during the reprogramming process. Furthermore, we reveal that OCT4 directly activates expression of Ash2l-b, and that ASH2L-B is a major isoform of ASH2L highly expressed in ESCs and required for somatic cell reprogramming. Together, this study emphasizes the importance of ubiquitination manipulation of the reprogramming factor and its interplay with the epigenetic regulator for successful reprogramming, opening a new avenue to improve the efficiency of pluripotency induction. Five lysine residues are major ubiquitination sites of OCT4 catalyzed by WWP2 Disruption of OCT4 ubiquitination promotes somatic cell reprogramming Mutation of OCT4 ubiquitination sites enhances OCT4 stability and H3K4me levels ASH2L-B contributes to somatic cell reprogramming as a direct target of OCT4
Collapse
Affiliation(s)
- Shuang Li
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Feng Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200032, China
| | - Junmei Zhang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaozhi Sun
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Han Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200032, China
| | - Yanwu Zeng
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Jing Hu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Fan Tang
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Junjie Gu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Yingming Zhao
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ying Jin
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200032, China.
| | - Bing Liao
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
32
|
Maiuthed A, Bhummaphan N, Luanpitpong S, Mutirangura A, Aporntewan C, Meeprasert A, Rungrotmongkol T, Rojanasakul Y, Chanvorachote P. Nitric oxide promotes cancer cell dedifferentiation by disrupting an Oct4:caveolin-1 complex: A new regulatory mechanism for cancer stem cell formation. J Biol Chem 2018; 293:13534-13552. [PMID: 29986880 DOI: 10.1074/jbc.ra117.000287] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 06/19/2018] [Indexed: 01/11/2023] Open
Abstract
Cancer stem cells (CSCs) are unique populations of cells that can self-renew and generate different cancer cell lineages. Although CSCs are believed to be a promising target for novel therapies, the specific mechanisms by which these putative therapeutics could intervene are less clear. Nitric oxide (NO) is a biological mediator frequently up-regulated in tumors and has been linked to cancer aggressiveness. Here, we search for targets of NO that could explain its activity. We find that it directly affects the stability and function of octamer-binding transcription factor 4 (Oct4), known to drive the stemness of lung cancer cells. We demonstrated that NO promotes the CSC-regulatory activity of Oct4 through a mechanism that involves complex formation between Oct4 and the scaffolding protein caveolin-1 (Cav-1). In the absence of NO, Oct4 forms a molecular complex with Cav-1, which promotes the ubiquitin-mediated proteasomal degradation of Oct4. NO promotes Akt-dependent phosphorylation of Cav-1 at tyrosine 14, disrupting the Cav-1:Oct4 complex. Site-directed mutagenesis and computational modeling studies revealed that the hydroxyl moiety at tyrosine 14 of Cav-1 is crucial for its interaction with Oct4. Both removal of the hydroxyl via mutation to phenylalanine and phosphorylation lead to an increase in binding free energy (ΔGbind) between Oct4 and Cav-1, destabilizing the complex. Together, these results unveiled a novel mechanism of CSC regulation through NO-mediated stabilization of Oct4, a key stem cell transcription factor, and point to new opportunities to design CSC-related therapeutics.
Collapse
Affiliation(s)
- Arnatchai Maiuthed
- From the Department of Pharmacology and Physiology.,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences
| | - Narumol Bhummaphan
- Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences.,the Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sudjit Luanpitpong
- the Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700 Thailand, and
| | - Apiwat Mutirangura
- the Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, and
| | | | - Arthitaya Meeprasert
- Structural and Computational Biology Research Group, and Department of Biochemistry, Faculty of Science
| | - Thanyada Rungrotmongkol
- Structural and Computational Biology Research Group, and Department of Biochemistry, Faculty of Science.,Ph.D. Program in Bioinformatics and Computational Biology
| | - Yon Rojanasakul
- WVU Cancer Institute, West Virginia University, Morgantown, West Virginia 26506
| | - Pithi Chanvorachote
- From the Department of Pharmacology and Physiology, .,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences
| |
Collapse
|
33
|
Choi J, Baek KH. Cellular functions of stem cell factors mediated by the ubiquitin-proteasome system. Cell Mol Life Sci 2018; 75:1947-1957. [PMID: 29423528 PMCID: PMC11105287 DOI: 10.1007/s00018-018-2770-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/12/2018] [Accepted: 02/01/2018] [Indexed: 12/16/2022]
Abstract
Stem cells undergo partitioning through mitosis and separate into specific cells of each of the three embryonic germ layers: endoderm, mesoderm, and ectoderm. Pluripotency, reprogramming, and self-renewal are essential elements of embryonic stem cells (ESCs), and it is becoming evident that regulation of protein degradation mediated by the ubiquitin-proteasome system (UPS) is one of the key cellular mechanisms in ESCs. Although the framework of that mechanism may seem simple, it involves complicated proteolytic machinery. The UPS controls cell development, survival, differentiation, lineage commitment, migration, and homing processes. This review is centered on the connection between stem cell factors NANOG, OCT-3/4, SOX2, KLF4, C-MYC, LIN28, FAK, and telomerase and the UPS. Herein, we summarize recent findings and discuss potential UPS mechanisms involved in pluripotency, reprogramming, differentiation, and self-renewal. Interactions between the UPS and stem cell transcription factors can apply to various human diseases which can be treated by generating more efficient iPSCs. Such complexes may permit the design of novel therapeutics and the establishment of biomarkers that may be used in diagnosis and prognosis development. Therefore, the UPS is an important target for stem cell therapeutic product research.
Collapse
Affiliation(s)
- Jihye Choi
- Department of Biomedical Science, CHA Stem Cell Institute, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA Stem Cell Institute, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea.
| |
Collapse
|
34
|
Li Z, Wang Y, Li Y, Yin W, Mo L, Qian X, Zhang Y, Wang G, Bu F, Zhang Z, Ren X, Zhu B, Niu C, Xiao W, Zhang W. Ube2s stabilizes β-Catenin through K11-linked polyubiquitination to promote mesendoderm specification and colorectal cancer development. Cell Death Dis 2018; 9:456. [PMID: 29674637 PMCID: PMC5908793 DOI: 10.1038/s41419-018-0451-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 01/01/2023]
Abstract
The canonical Wnt/β-Catenin signaling pathway is widely involved in regulating diverse biological processes. Dysregulation of the pathway results in severe consequences, such as developmental defects and malignant cancers. Here, we identified Ube2s as a novel activator of the Wnt/β-Catenin signaling pathway. It modified β-Catenin at K19 via K11-linked polyubiquitin chain. This modification resulted in an antagonistic effect against the destruction complex/β-TrCP cascade-orchestrated β-Catenin degradation. As a result, the stability of β-Catenin was enhanced, thus promoting its cellular accumulation. Importantly, Ube2s-promoted β-Catenin accumulation partially released the dependence on exogenous molecules for the process of embryonic stem (ES) cell differentiation into mesoendoderm lineages. Moreover, we demonstrated that UBE2S plays a critical role in determining the malignancy properties of human colorectal cancer (CRC) cells in vitro and in vivo. The findings in this study extend our mechanistic understanding of the mesoendodermal cell fate commitment, and provide UBE2S as a putative target for human CRC therapy.
Collapse
Affiliation(s)
- Zhaoyan Li
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Yan Wang
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Yadan Li
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Wanqi Yin
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Libin Mo
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Xianghao Qian
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Yiran Zhang
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Guifen Wang
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Fan Bu
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Zhiling Zhang
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Xiaofang Ren
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Baochang Zhu
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Chang Niu
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Wei Xiao
- College of Life Sciences, Capital Normal University, Beijing, China.
| | - Weiwei Zhang
- College of Life Sciences, Capital Normal University, Beijing, China.
| |
Collapse
|
35
|
Post-translational modification of OCT4 in breast cancer tumorigenesis. Cell Death Differ 2018; 25:1781-1795. [PMID: 29511337 PMCID: PMC6180041 DOI: 10.1038/s41418-018-0079-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 01/08/2018] [Accepted: 01/24/2018] [Indexed: 01/06/2023] Open
Abstract
Recurrence and drug resistance of breast cancer are still the main reasons for breast cancer-associated deaths. Cancer stem cell (CSC) model has been proposed as a hypothesis for the lethality of breast cancer. Molecular mechanisms underlying CSC maintenance are still unclear. In this study, we generated mammospheres derived from breast cancer MDA-MB231 cells and MCF7 cells to enrich CSCs and performed DNA microarray analysis. We found that the expression of carboxy terminus of HSP70-interacting protein (CHIP) E3 ubiquitin ligase was significantly downregulated in breast CSCs. CHIP depletion increased mammosphere formation, whereas CHIP overexpression reversed this effect. We identified interactomes by mass spectrometry and detected CHIP directly interacted with OCT4, a stemness factor. CHIP overexpression decreased OCT4 stability through proteasomal degradation. CHIP induced OCT4 ubiquitination, whereas H260Q, a catalytic CHIP mutant, did not. Interestingly, we determined that OCT4 was ubiquitinated at lysine 284, and CHIP overexpression did not degrade K284R mutant OCT4. CHIP overexpression decreased the proliferation and side population of breast cancer cells, but these were not occurred in K284R mutant OCT4 overexpressed cells. Only 1000 cells showing CHIP depletion or OCT4 overexpression sufficiently generated breast tumors and lung metastases in xenografted mice. Ubiquitination-defective mutant of OCT4(K284R) overexpressed cells drastically generated tumor burdens in mice. Patients with breast cancer who showed low CHIP expression had poor survival probability. Taken together, we suggest that CHIP-induced OCT4 ubiquitination is important in breast CSCs. Regulation of CHIP expression and OCT4 protein stability is a considerable approach for breast cancer therapy.
Collapse
|
36
|
Zou G, Liu T, Guo L, Huang Y, Feng Y, Duan T. MicroRNA‑32 silences WWP2 expression to maintain the pluripotency of human amniotic epithelial stem cells and β islet‑like cell differentiation. Int J Mol Med 2018; 41:1983-1991. [PMID: 29393344 PMCID: PMC5810217 DOI: 10.3892/ijmm.2018.3436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/20/2017] [Indexed: 11/06/2022] Open
Abstract
Human amniotic epithelial stem cells (HuAECs) exhibit pluripotent characteristics, which are similar to those of embryonic stem cells, and can differentiate into various adult tissues and cells through directed induction. However, in culture, HuAECs tend to lose their pluripotency, and their directed differentiation capability declines with increasing passage number. The stem cell pluripotency factor octamer-binding protein 4 (Oct4) is an important transcription factor that promotes stem cell self-proliferation and maintains their pluripotency. Previous studies have demonstrated that WW domain containing E3 ubiquitin protein ligase 2 (WWP2) negatively regulates Oct4 expression and stem cell pluripotency. Therefore, the present study aimed to investigate the regulation of WWP2 by microRNAs (miRs), and to evaluate the expression of the downstream factor Oct4 and the maintenance of HuAEC pluripotency. Bioinformatics analysis identified a complementary binding site for miR-32 in the 3′untranslated region of the WWP2 gene, thus suggesting that it may be a target gene of miR-32. Post-infection of HuAECs with a vector overexpressing miR-32, the endogenous expression of WWP2 was significantly decreased, whereas Oct4 expression was significantly increased. Furthermore, miR-32-infected cells differentiated into β islet-like cells by directed induction. The results indicated that after induction, HuAECs overexpressing miR-32 also overexpressed the biomarkers of β islet-like cells. In addition, the ability to secrete insulin was markedly enhanced in response to glucose stimulation, in cells overexpressing miR-32. In conclusion, the present study suggested that miR-32 may effectively inhibit WWP2 expression in HuAECs and promote Oct4 overexpression to maintain their pluripotency.
Collapse
Affiliation(s)
- Gang Zou
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, P.R. China
| | - Te Liu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, P.R. China
| | - Lihe Guo
- Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Yongyi Huang
- Laboratoire PROTEE, Batiment R, University du Sud Toulon‑Var, 83957 La Garde Cedex, France
| | - Ya Feng
- Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200040, P.R. China
| | - Tao Duan
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, P.R. China
| |
Collapse
|
37
|
Yu Z, Li T, Wang C, Deng S, Zhang B, Huo X, Zhang B, Wang X, Zhong Y, Ma X. Gamabufotalin triggers c-Myc degradation via induction of WWP2 in multiple myeloma cells. Oncotarget 2017; 7:15725-37. [PMID: 26894970 PMCID: PMC4941272 DOI: 10.18632/oncotarget.7398] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/05/2016] [Indexed: 12/14/2022] Open
Abstract
Deciding appropriate therapy for multiple myeloma (MM) is challenging because of the occurrence of multiple chromosomal changes and the fatal nature of the disease. In the current study, gamabufotalin (GBT) was isolated from toad venom, and its tumor-specific cytotoxicity was investigated in human MM cells. We found GBT inhibited cell growth and induced apoptosis with the IC50 values <50 nM. Mechanistic studies using functional approaches identified GBT as an inhibitor of c-Myc. Further analysis showed that GBT especially evoked the ubiquitination and degradation of c-Myc protein, thereby globally repressing the expression of c-Myc target genes. GBT treatment inhibited ERK and AKT signals, while stimulating the activation of JNK cascade. An E3 ubiquitin-protein ligase, WWP2, was upregulated following JNK activation and played an important role in c-Myc ubiquitination and degradation through direct protein-protein interaction. The antitumor effect of GBT was validated in a xenograft mouse model and the suppression of MM-induced osteolysis was verified in a SCID-hu model in vivo. Taken together, our study identified the potential of GBT as a promising therapeutic agent in the treatment of MM.
Collapse
Affiliation(s)
- Zhenlong Yu
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Tao Li
- Department of Biology, College of Chemistry and Life Sciences, Zhejiang Normal University, Zhejiang, China
| | - Chao Wang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Sa Deng
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Baojing Zhang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiaokui Huo
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaobo Wang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yuping Zhong
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaochi Ma
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
38
|
He M, Zhou Z, Wu G, Chen Q, Wan Y. Emerging role of DUBs in tumor metastasis and apoptosis: Therapeutic implication. Pharmacol Ther 2017; 177:96-107. [PMID: 28279784 PMCID: PMC5565705 DOI: 10.1016/j.pharmthera.2017.03.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Malfunction of ubiquitin-proteasome system is tightly linked to tumor formation and tumor metastasis. Targeting the ubiquitin-pathway provides a new strategy for anti-cancer therapy. Despite the parts played by ubiquitin modifiers, removal of ubiquitin from the functional proteins by the deubiquitinating enzymes (DUBs) plays an important role in governing the multiple steps of the metastatic cascade, including local invasion, dissemination, and eventual colonization of the tumor to distant organs. Both deregulated ubiquitination and deubiquitination could lead to dysregulation of various critical events and pathways such as apoptosis and epithelial-mesenchymal transition (EMT). Recent TCGA study has further revealed the connection between mutations of DUBs and various types of tumors. In addition, emerging drug design targeting DUBs provides a new strategy for anti-cancer therapy. In this review, we will summarize the role of deubiquitination and highlight the recent discoveries of DUBs with regards to multiple metastatic events including anti-apoptosis pathway and EMT. We will further discuss the regulation of deubiquitination as a novel strategy for anti-cancer therapy.
Collapse
Affiliation(s)
- Mingjing He
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Zhuan Zhou
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - George Wu
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Yong Wan
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
39
|
Werner A, Manford AG, Rape M. Ubiquitin-Dependent Regulation of Stem Cell Biology. Trends Cell Biol 2017; 27:568-579. [PMID: 28528988 PMCID: PMC5643009 DOI: 10.1016/j.tcb.2017.04.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 04/04/2017] [Accepted: 04/10/2017] [Indexed: 12/21/2022]
Abstract
The growth of a metazoan body relies on a series of highly coordinated cell-fate decisions by stem cells which can undergo self-renewal, reversibly enter a quiescent state, or terminally commit to a cell specification program. To guide their decisions, stem cells make frequent use of ubiquitylation, a post-translational modification that can affect the activity, interaction landscape, or stability of stem cell proteins. In this review we discuss novel findings that have provided insight into ubiquitin-dependent mechanisms of stem cell control and revealed how an essential and highly conserved protein modification can shape metazoan development.
Collapse
Affiliation(s)
- Achim Werner
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Andrew G Manford
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Michael Rape
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
40
|
French ME, Klosowiak JL, Aslanian A, Reed SI, Yates JR, Hunter T. Mechanism of ubiquitin chain synthesis employed by a HECT domain ubiquitin ligase. J Biol Chem 2017; 292:10398-10413. [PMID: 28461335 DOI: 10.1074/jbc.m117.789479] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 04/27/2017] [Indexed: 11/06/2022] Open
Abstract
Homologous to E6AP C-terminal (HECT) ubiquitin (Ub) ligases (E3s) are a large class of enzymes that bind to their substrates and catalyze ubiquitination through the formation of a Ub thioester intermediate. The mechanisms by which these E3s assemble polyubiquitin chains on their substrates remain poorly defined. We report here that the Nedd4 family HECT E3, WWP1, assembles substrate-linked Ub chains containing Lys-63, Lys-48, and Lys-11 linkages (Lys-63 > Lys-48 > Lys-11). Our results demonstrate that WWP1 catalyzes the formation of Ub chains through a sequential addition mechanism, in which Ub monomers are transferred in a successive fashion to the substrate, and that ubiquitination by WWP1 requires the presence of a low-affinity, noncovalent Ub-binding site within the HECT domain. Unexpectedly, we find that the formation of Ub chains by WWP1 occurs in two distinct phases. In the first phase, chains are synthesized in a unidirectional manner and are linked exclusively through Lys-63 of Ub. In the second phase, chains are elongated in a multidirectional fashion characterized by the formation of mixed Ub linkages and branched structures. Our results provide new insight into the mechanism of Ub chain formation employed by Nedd4 family HECT E3s and suggest a framework for understanding how this family of E3s generates Ub signals that function in proteasome-independent and proteasome-dependent pathways.
Collapse
Affiliation(s)
- Michael E French
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037.,the Departments of Cell and Molecular Biology and
| | - Julian L Klosowiak
- the Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Aaron Aslanian
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037.,Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, and
| | | | - John R Yates
- Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, and
| | - Tony Hunter
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037,
| |
Collapse
|
41
|
Effects of MG132 on the in vitro development and epigenetic modification of Debao porcine somatic cell nuclear transfer embryos. Theriogenology 2017; 94:48-58. [DOI: 10.1016/j.theriogenology.2017.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/30/2016] [Accepted: 02/03/2017] [Indexed: 01/12/2023]
|
42
|
Fütterer A, de Celis J, Navajas R, Almonacid L, Gutiérrez J, Talavera-Gutiérrez A, Pacios-Bras C, Bernascone I, Martin-Belmonte F, Martinéz-A C. DIDO as a Switchboard that Regulates Self-Renewal and Differentiation in Embryonic Stem Cells. Stem Cell Reports 2017; 8:1062-1075. [PMID: 28330622 PMCID: PMC5390109 DOI: 10.1016/j.stemcr.2017.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 12/28/2022] Open
Abstract
Transition from symmetric to asymmetric cell division requires precise coordination of differential gene expression. We show that embryonic stem cells (ESCs) mainly express DIDO3 and that their differentiation after leukemia inhibitory factor withdrawal requires DIDO1 expression. C-terminal truncation of DIDO3 (Dido3ΔCT) impedes ESC differentiation while retaining self-renewal; small hairpin RNA-Dido1 ESCs have the same phenotype. Dido3ΔCT ESC differentiation is rescued by ectopic expression of DIDO3, which binds the Dido locus via H3K4me3 and RNA POL II and induces DIDO1 expression. DIDO1, which is exported to cytoplasm, associates with, and is N-terminally phosphorylated by PKCiota. It binds the E3 ubiquitin ligase WWP2, which contributes to cell fate by OCT4 degradation, to allow expression of primitive endoderm (PE) markers. PE formation also depends on phosphorylated DIDO3 localization to centrosomes, which ensures their correct positioning for PE cell polarization. We propose that DIDO isoforms act as a switchboard that regulates genetic programs for ESC transition from pluripotency maintenance to promotion of differentiation. DIDO3 regulates DIDO1 expression Cytoplasmic DIDO1 promotes cell fate DIDO3 regulates centrosome position DIDO1 and DIDO3 are phosphorylated by PKCiota
Collapse
Affiliation(s)
- Agnes Fütterer
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Jésus de Celis
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Rosana Navajas
- Proteomics Unit, ProteoRed ISCIII, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Luis Almonacid
- Genomics Unit, Q-PCR Facility, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Julio Gutiérrez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Amaia Talavera-Gutiérrez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Cristina Pacios-Bras
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Ilenia Bernascone
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Fernando Martin-Belmonte
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Carlos Martinéz-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain.
| |
Collapse
|
43
|
Wen J, Zeng Y, Fang Z, Gu J, Ge L, Tang F, Qu Z, Hu J, Cui Y, Zhang K, Wang J, Li S, Sun Y, Jin Y. Single-cell analysis reveals lineage segregation in early post-implantation mouse embryos. J Biol Chem 2017; 292:9840-9854. [PMID: 28298438 DOI: 10.1074/jbc.m117.780585] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/13/2017] [Indexed: 11/06/2022] Open
Abstract
The mammalian post-implantation embryo has been extensively investigated at the tissue level. However, to unravel the molecular basis for the cell-fate plasticity and determination, it is essential to study the characteristics of individual cells. In particular, the individual definitive endoderm (DE) cells have not been characterized in vivo Here, we report gene expression patterns in single cells freshly isolated from mouse embryos on days 5.5 and 6.5. Initial transcriptome data from 124 single cells yielded signature genes for the epiblast, visceral endoderm, and extra-embryonic ectoderm and revealed a unique distribution pattern of fibroblast growth factor (FGF) ligands and receptors. Further analysis indicated that early-stage epiblast cells do not segregate into lineages of the major germ layers. Instead, some cells began to diverge from epiblast cells, displaying molecular features of the premesendoderm by expressing higher levels of mesendoderm markers and lower levels of Sox3 transcripts. Analysis of single-cell high-throughput quantitative RT-PCR data from 441 cells identified a late stage of the day 6.5 embryo in which mesoderm and DE cells emerge, with many of them coexpressing Oct4 and Gata6 Analysis of single-cell RNA-sequence data from 112 cells of the late-stage day 6.5 embryos revealed differentially expressed signaling genes and networks of transcription factors that might underlie the segregation of the mesoderm and DE lineages. Moreover, we discovered a subpopulation of mesoderm cells that possess molecular features of the extraembryonic mesoderm. This study provides fundamental insight into the molecular basis for lineage segregation in post-implantation mouse embryos.
Collapse
Affiliation(s)
- Jing Wen
- From the Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031
| | - Yanwu Zeng
- the Department of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, and
| | - Zhuoqing Fang
- From the Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031
| | - Junjie Gu
- the Department of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, and
| | - Laixiang Ge
- the Department of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, and
| | - Fan Tang
- the Department of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, and
| | - Zepeng Qu
- the Department of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, and
| | - Jing Hu
- the Department of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, and
| | - Yaru Cui
- the Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Kushan Zhang
- the Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Junbang Wang
- the Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Siguang Li
- the Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Yi Sun
- the Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Ying Jin
- From the Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, .,the Department of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, and
| |
Collapse
|
44
|
Kwon SK, Lee DH, Kim SY, Park JH, Choi J, Baek KH. Ubiquitin-specific protease 21 regulating the K48-linked polyubiquitination of NANOG. Biochem Biophys Res Commun 2016; 482:1443-1448. [PMID: 27956178 DOI: 10.1016/j.bbrc.2016.12.055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 10/20/2022]
Abstract
NANOG, one of homeobox proteins, plays a crucial role in regulating self-renewal and pluripotency for embryonic stem cells (ESCs). Since the ubiquitin-mediated degradation of NANOG protein has been implicated in its cellular functions involved in not only maintenance of pluripotency and pluripotent epiblast, but also prevention of primitive endoderm differentiation, the identification of ubiquitin ligases and deubiquitinating enzymes (DUBs) for NANOG is required to elucidate its protein stability and the regulation of cellular functions in these processes. In this study, we have identified a novel deubiquitinating enzyme USP21 which interacts with NANOG by both yeast two hybrid screening for DUBs and immunoprecipitation analyses. These analyses revealed that USP21 specifically regulates the Lys48-linked polyubiquitination and stability of NANOG, providing a new way of maintaining the pluripotency of ESCs and induced pluripotent stem cells (iPSCs).
Collapse
Affiliation(s)
- Seul-Ki Kwon
- Department of Biomedical Science, CHA University, Gyeonggi-Do, 13488, Republic of Korea
| | - Da-Hye Lee
- Department of Biomedical Science, CHA University, Gyeonggi-Do, 13488, Republic of Korea
| | - Soo-Yeon Kim
- Department of Biomedical Science, CHA University, Gyeonggi-Do, 13488, Republic of Korea
| | - Jung-Hyun Park
- Department of Biomedical Science, CHA University, Gyeonggi-Do, 13488, Republic of Korea
| | - Jihye Choi
- Department of Biomedical Science, CHA University, Gyeonggi-Do, 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do, 13488, Republic of Korea.
| |
Collapse
|
45
|
Liu J, Wan L, Liu J, Yuan Z, Zhang J, Guo J, Malumbres M, Liu J, Zou W, Wei W. Cdh1 inhibits WWP2-mediated ubiquitination of PTEN to suppress tumorigenesis in an APC-independent manner. Cell Discov 2016; 2:15044. [PMID: 27462441 PMCID: PMC4860961 DOI: 10.1038/celldisc.2015.44] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/05/2015] [Indexed: 02/05/2023] Open
Abstract
Anaphase-promoting complex/cyclosome/Cdh1 is a multi-subunit ubiquitin E3 ligase that drives M to G1 cell cycle progression through primarily earmarking various substrates for ubiquitination and subsequent degradation by the 26S proteasome. Notably, emerging evidence suggested that Cdh1 could also function in various cellular processes independent of anaphase-promoting complex/cyclosome. To this end, we recently identified an anaphase-promoting complex/cyclosome-independent function of Cdh1 in modulating osteoblast differentiation through activating Smurf1, one of the NEDD4 family of HECT domain-containing E3 ligases. However, it remains largely unknown whether Cdh1 could exert its tumor suppressor role through similarly modulating the E3 ligase activities of other NEDD4 family members, most of which have characterized important roles in tumorigenesis. Here we report that in various tumor cells, Cdh1, conversely, suppresses the E3 ligase activity of WWP2, another NEDD4 family protein, in an anaphase-promoting complex/cyclosome-independent manner. As such, loss of Cdh1 activates WWP2, leading to reduced abundance of WWP2 substrates including PTEN, which subsequently activates PI3K/Akt oncogenic signaling to facilitate tumorigenesis. This study expands the non-anaphase-promoting complex/cyclosome function of Cdh1 in regulating the NEDD4 family E3 ligases, and further suggested that enhancing Cdh1 to inhibit the E3 ligase activity of WWP2 could be a promising strategy for treating human cancers.
Collapse
Affiliation(s)
- Jia Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Life Science, FIST, Xi'an Jiaotong University, Xi'an, China; Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lixin Wan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA, USA
| | - Jing Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Life Science, FIST, Xi'an Jiaotong University, Xi'an, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhu Yuan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; State Key Laboratory of Biotherapy and Cancer Center, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinfang Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA, USA
| | - Jianfeng Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Marcos Malumbres
- Cell Division and Cancer group, Spanish National Cancer Research Centre (CNIO) , Madrid, Spain
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Life Science, FIST, Xi'an Jiaotong University , Xi'an, China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA, USA
| |
Collapse
|
46
|
Matic I, Antunovic M, Brkic S, Josipovic P, Mihalic KC, Karlak I, Ivkovic A, Marijanovic I. Expression of OCT-4 and SOX-2 in Bone Marrow-Derived Human Mesenchymal Stem Cells during Osteogenic Differentiation. Open Access Maced J Med Sci 2016; 4:9-16. [PMID: 27275321 PMCID: PMC4884261 DOI: 10.3889/oamjms.2016.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 11/17/2022] Open
Abstract
AIM: Determine the levels of expression of pluripotency genes OCT-4 and SOX-2 before and after osteogenic differentiation of human mesenchymal stem cells (hMSCs). METHODS: Human MSCs were derived from the bone marrow and differentiated into osteoblasts. The analyses were performed on days 0 and 14 of the cell culture. In vitro differentiation was evaluated due to bone markers – alkaline phosphatase (AP) activity and the messenger RNA (mRNA) expression of AP and bone sialoprotein (BSP). The OCT-4 and SOX-2 expression was evaluated at mRNA level by real-time qPCR and at protein level by immunocytochemistry. RESULTS: In vitro cultures on day 14 showed an increase in AP activity and upregulation of AP and BSP gene expression. OCT-4 and SOX-2 in undifferentiated hMSCs on day 0 is detectable and very low compared to tumor cell lines as a positive control. Immunocytochemistry detected OCT-4 in the cell nuclei prior (day 0) and post differentiation (day 14). On the same time points, cultures were negative for SOX-2 protein. CONCLUSION: Messenger RNA for pluripotency markers OCT-4 and SOX-2 isolated from hMSCs was less present, while OCT-4 protein was detected in cell nuclei prior and post differentiation into osteoblast lineage.
Collapse
Affiliation(s)
- Igor Matic
- Division of Biology, Department of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Maja Antunovic
- Division of Biology, Department of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Sime Brkic
- Division of Biology, Department of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Pavle Josipovic
- Division of Biology, Department of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Katarina Caput Mihalic
- Division of Biology, Department of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Ivan Karlak
- Department of Traumatology, University Hospital Sestre Milosrdnice, Zagreb, Croatia
| | | | - Inga Marijanovic
- Division of Biology, Department of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
47
|
The Importance of Ubiquitination and Deubiquitination in Cellular Reprogramming. Stem Cells Int 2016; 2016:6705927. [PMID: 26880980 PMCID: PMC4736574 DOI: 10.1155/2016/6705927] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/04/2015] [Accepted: 11/12/2015] [Indexed: 01/09/2023] Open
Abstract
Ubiquitination of core stem cell transcription factors can directly affect stem cell maintenance and differentiation. Ubiquitination and deubiquitination must occur in a timely and well-coordinated manner to regulate the protein turnover of several stemness related proteins, resulting in optimal embryonic stem cell maintenance and differentiation. There are two switches: an E3 ubiquitin ligase enzyme that tags ubiquitin molecules to the target proteins for proteolysis and a second enzyme, the deubiquitinating enzyme (DUBs), that performs the opposite action, thereby preventing proteolysis. In order to maintain stemness and to allow for efficient differentiation, both ubiquitination and deubiquitination molecular switches must operate properly in a balanced manner. In this review, we have summarized the importance of the ubiquitination of core stem cell transcription factors, such as Oct3/4, c-Myc, Sox2, Klf4, Nanog, and LIN28, during cellular reprogramming. Furthermore, we emphasize the role of DUBs in regulating core stem cell transcriptional factors and their function in stem cell maintenance and differentiation. We also discuss the possibility of using DUBs, along with core transcription factors, to efficiently generate induced pluripotent stem cells. Our review provides a relatively new understanding regarding the importance of ubiquitination/deubiquitination of stem cell transcription factors for efficient cellular reprogramming.
Collapse
|
48
|
Qian X, Kim JK, Tong W, Villa-Diaz LG, Krebsbach PH. DPPA5 Supports Pluripotency and Reprogramming by Regulating NANOG Turnover. Stem Cells 2015; 34:588-600. [PMID: 26661329 DOI: 10.1002/stem.2252] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 09/19/2015] [Accepted: 10/18/2015] [Indexed: 01/16/2023]
Abstract
Although a specific group of transcription factors such as OCT4, SOX2, and NANOG are known to play essential roles in pluripotent stem cell (PSC) self-renewal, pluripotency, and reprogramming, other factors and the key signaling pathways regulating these important properties are not completely understood. Here, we demonstrate that the PSC marker Developmental Pluripotency Associated 5 (DPPA5) plays an important role in human PSC (hPSC) self-renewal and cell reprogramming in feeder-free conditions. Compared to hPSCs grown on mouse embryonic fibroblasts, cells cultured on feeder-free substrates, such as Matrigel, Laminin-511, Vitronectin, or the synthetic polymer poly[2-(methacryloyloxy) ethyl dimethyl-(3-sulfopropyl) ammonium hydroxide], had significantly higher DPPA5 gene expression and protein levels. Overexpression of DPPA5 in hPSCs increased NANOG protein levels via a post-transcriptional mechanism. Coimmunoprecipitation, protein stability assays, and quantitative RT-PCR, demonstrated that DPPA5 directly interacted, stabilized, and enhanced the function of NANOG in hPSCs. Additionally, DPPA5 increased the reprogramming efficiency of human somatic cells to induced pluripotent stem cells (hiPSCs). Our study provides new insight into the function of DPPA5 and NANOG regulation in hPSCs.
Collapse
Affiliation(s)
- Xu Qian
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Jin Koo Kim
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Wilbur Tong
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Luis G Villa-Diaz
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul H Krebsbach
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
49
|
Liu C, Zhang D, Shen Y, Tao X, Liu L, Zhong Y, Fang S. DPF2 regulates OCT4 protein level and nuclear distribution. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:3279-3293. [PMID: 26417682 DOI: 10.1016/j.bbamcr.2015.09.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/28/2015] [Accepted: 09/21/2015] [Indexed: 02/05/2023]
Abstract
The amount of transcription factor OCT4 is strictly regulated. A tight regulation of OCT4 levels is crucial for mammalian embryonic development and oncogenesis. However, the mechanisms underlying regulation of OCT4 protein expression and nuclear distribution are largely unknown. Here, we report that DPF2, a plant homeodomain (PHD) finger protein, is upregulated during H9 cell differentiation induced by retinoic acid. Endogenous interaction between DPF2 and OCT4 in P19 cells was revealed by an immunoprecipitation assay. GST-pull down assay proved that OCT4 protein in H9 cells and recombinant OCT4 can precipitate with DPF2 in vitro. In vitro ubiquitination assay demonstrated DPF2 might serve as an E3 ligase. Knock down of dpf2 using siRNA increased OCT4 protein level and stability in P19 cells. DPF2 siRNAs also up-regulates OCT4 but not NANOG in H9 cells. However, RA fails to downregulates OCT4 protein level in cells infected by lenitviruses containing DPF2 siRNA. Moreover, overexpression of both DPF2 and OCT4 in 293 cells proved the DPF2-OCT4 interaction. DPF2 but not PHD2 mutant DPF2 enhanced ubiquitination and degradation of OCT4 in 293 cells co-expressed DPF2 and OCT4. Both wild type DPF2 and PHD2 mutant DPF2 redistributes nuclear OCT4 without affecting DPF2-OCT4 interaction. Further analysis indicated that DPF2 decreases monomeric and mono-ubiquitinated OCT4, assembles poly-ubiquitin chains on OCT4 mainly through Ub-K48 linkage. These findings contribute to an understanding of how OCT4 protein level and nuclear distribution is regulated by its associated protein.
Collapse
Affiliation(s)
- Chao Liu
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032 China; Center for Biomedical Engineering and Technology (BioMET), University of Maryland, Baltimore, MD 21201 USA.
| | - Dijuan Zhang
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032 China
| | - Yuxian Shen
- School of Basic Medical Sciences, Institute of Biopharmaceuticals, Anhui Medical University, Hefei, Anhui 230032 China
| | - Xiaofang Tao
- School of Basic Medical Sciences, Institute of Biopharmaceuticals, Anhui Medical University, Hefei, Anhui 230032 China
| | - Lihua Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yongwang Zhong
- Center for Biomedical Engineering and Technology (BioMET), University of Maryland, Baltimore, MD 21201 USA
| | - Shengyun Fang
- Center for Biomedical Engineering and Technology (BioMET), University of Maryland, Baltimore, MD 21201 USA.
| |
Collapse
|
50
|
Bahnassawy L, Perumal TM, Gonzalez-Cano L, Hillje AL, Taher L, Makalowski W, Suzuki Y, Fuellen G, del Sol A, Schwamborn JC. TRIM32 modulates pluripotency entry and exit by directly regulating Oct4 stability. Sci Rep 2015; 5:13456. [PMID: 26307407 PMCID: PMC4642535 DOI: 10.1038/srep13456] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 07/17/2015] [Indexed: 12/27/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have revolutionized the world of regenerative medicine; nevertheless, the exact molecular mechanisms underlying their generation and differentiation remain elusive. Here, we investigated the role of the cell fate determinant TRIM32 in modulating such processes. TRIM32 is essential for the induction of neuronal differentiation of neural stem cells by poly-ubiquitinating cMyc to target it for degradation resulting in inhibition of cell proliferation. To elucidate the role of TRIM32 in regulating somatic cell reprogramming we analysed the capacity of TRIM32-knock-out mouse embryonic fibroblasts (MEFs) in generating iPSC colonies. TRIM32 knock-out MEFs produced a higher number of iPSC colonies indicating a role for TRIM32 in inhibiting this cellular transition. Further characterization of the generated iPSCs indicated that the TRIM32 knock-out iPSCs show perturbed differentiation kinetics. Additionally, mathematical modelling of global gene expression data revealed that during differentiation an Oct4 centred network in the wild-type cells is replaced by an E2F1 centred network in the TRIM32 deficient cells. We show here that this might be caused by a TRIM32-dependent downregulation of Oct4. In summary, the data presented here reveal that TRIM32 directly regulates at least two of the four Yamanaka Factors (cMyc and Oct4), to modulate cell fate transitions.
Collapse
Affiliation(s)
- Lamia'a Bahnassawy
- Westfälische Wilhelms-Universität Münster, ZMBE, Institute of Cell Biology, Stem Cell Biology and Regeneration Group, Von-Esmarch-Str. 56, 48149 Münster, Germany.,Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, 7 avenue des Hauts-Fourneaux, 4362 Esch-Belval, Luxembourg
| | - Thanneer M Perumal
- Luxembourg Centre for Systems Biomedicine (LCSB), Computational Biology, University of Luxembourg, 7 avenue des Hauts-Fourneaux, 4362 Esch-Belval, Luxembourg
| | - Laura Gonzalez-Cano
- Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, 7 avenue des Hauts-Fourneaux, 4362 Esch-Belval, Luxembourg
| | - Anna-Lena Hillje
- Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, 7 avenue des Hauts-Fourneaux, 4362 Esch-Belval, Luxembourg
| | - Leila Taher
- Institute for Biostatistics and Informatics in Medicine und Ageing Research, Rostock University Medical Centre, Ernst-Heydemann-Str. 8, 18057 Rostock, Germany
| | - Wojciech Makalowski
- Westfälische Wilhelms-Universität Münster, Institute of Bioinformatics, Niels-Stensen-Straße 14, 48149 Münster, Germany
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba-ken 227-8561, Japan
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine und Ageing Research, Rostock University Medical Centre, Ernst-Heydemann-Str. 8, 18057 Rostock, Germany
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), Computational Biology, University of Luxembourg, 7 avenue des Hauts-Fourneaux, 4362 Esch-Belval, Luxembourg
| | - Jens Christian Schwamborn
- Westfälische Wilhelms-Universität Münster, ZMBE, Institute of Cell Biology, Stem Cell Biology and Regeneration Group, Von-Esmarch-Str. 56, 48149 Münster, Germany.,Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, 7 avenue des Hauts-Fourneaux, 4362 Esch-Belval, Luxembourg
| |
Collapse
|