1
|
Li Y, Ma C, Sheng Y, Huang S, Sun H, Ti Y, Wang Z, Wang F, Chen F, Li C, Guo H, Tang M, Song F, Wang H, Zhong M. TRIB3 mediates vascular calcification by facilitating self-ubiquitination and dissociation of Smurf1 in chronic kidney disease. J Clin Invest 2025; 135:e175972. [PMID: 39932798 PMCID: PMC11957692 DOI: 10.1172/jci175972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The osteogenic environment promotes vascular calcium phosphate deposition and aggregation of unfolded and misfolded proteins, resulting in ER stress in chronic kidney disease (CKD). Controlling ER stress through genetic intervention is a promising approach for treating vascular calcification. In this study, we demonstrated a positive correlation between ER stress-induced tribble homolog 3 (TRIB3) expression and progression of vascular calcification in human and rodent CKD. Increased TRIB3 expression promoted vascular smooth muscle cell (VSMC) calcification by interacting with the C2 domain of the E3 ubiquitin-protein ligase Smurf1, facilitating its K48-related self-ubiquitination at Lys381 and Lys383 and subsequent dissociation from the plasma membrane and nuclei. This degeneration of Smurf1 accelerated the stabilization of the osteogenic transcription factors RUNX family transcription factor 2 (Runx2) and SMAD family member 1 (Smad1). C/EBP homologous protein and activating transcription factor 4 are upstream transcription factors of TRIB3 in an osteogenic environment. Genetic KO of TRIB3 or rescue of Smurf1 ameliorated VSMC and vascular calcification by stabilizing Smurf1 and enhancing the degradation of Runx2 and Smad1. Our findings shed light on the vital role of TRIB3 as a scaffold in ER stress and vascular calcification and offer a potential therapeutic option for CKD.
Collapse
Affiliation(s)
- Yihui Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Chang Ma
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yanan Sheng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shanying Huang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Huaibing Sun
- Department of Organ Transplantation, Qilu Hospital, and
| | - Yun Ti
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhihao Wang
- Department of Geriatric Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Wang
- Department of Critical Care Medicine, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Fangfang Chen
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Chen Li
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Haipeng Guo
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Mengxiong Tang
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China
| | - Fangqiang Song
- Department of Critical Care Medicine, Affiliated Tengzhou Hospital of Xuzhou Medical University/Tengzhou Central People’s Hospital, Shandong, China
| | - Hao Wang
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Ming Zhong
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
Zhang HR, Wang YH, Xiao ZP, Yang G, Xu YR, Huang ZT, Wang WZ, He F. E3 ubiquitin ligases: key regulators of osteogenesis and potential therapeutic targets for bone disorders. Front Cell Dev Biol 2024; 12:1447093. [PMID: 39211390 PMCID: PMC11358089 DOI: 10.3389/fcell.2024.1447093] [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: 06/11/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Ubiquitination is a crucial post-translational modification of proteins that mediates the degradation or functional regulation of specific proteins. This process participates in various biological processes such as cell growth, development, and signal transduction. E3 ubiquitin ligases play both positive and negative regulatory roles in osteogenesis and differentiation by ubiquitination-mediated degradation or stabilization of transcription factors, signaling molecules, and cytoskeletal proteins. These activities affect the proliferation, differentiation, survival, and bone formation of osteoblasts (OBs). In recent years, advances in genomics, transcriptomics, and proteomics have led to a deeper understanding of the classification, function, and mechanisms of action of E3 ubiquitin ligases. This understanding provides new insights and approaches for revealing the molecular regulatory mechanisms of bone formation and identifying therapeutic targets for bone metabolic diseases. This review discusses the research progress and significance of the positive and negative regulatory roles and mechanisms of E3 ubiquitin ligases in the process of osteogenic differentiation. Additionally, the review highlights the role of E3 ubiquitin ligases in bone-related diseases. A thorough understanding of the role and mechanisms of E3 ubiquitin ligases in osteogenic differentiation could provide promising therapeutic targets for bone tissue engineering based on stem cells.
Collapse
Affiliation(s)
- Heng-Rui Zhang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Yang-Hao Wang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhen-Ping Xiao
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
- Department of Pain and Rehabilitation, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Guang Yang
- Department of Trauma Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yun-Rong Xu
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Zai-Tian Huang
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Wei-Zhou Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fei He
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| |
Collapse
|
3
|
Gawden-Bone CM, Lehner PJ, Volkmar N. As a matter of fat: Emerging roles of lipid-sensitive E3 ubiquitin ligases. Bioessays 2023; 45:e2300139. [PMID: 37890275 DOI: 10.1002/bies.202300139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
The dynamic structure and composition of lipid membranes need to be tightly regulated to control the vast array of cellular processes from cell and organelle morphology to protein-protein interactions and signal transduction pathways. To maintain membrane integrity, sense-and-response systems monitor and adjust membrane lipid composition to the ever-changing cellular environment, but only a relatively small number of control systems have been described. Here, we explore the emerging role of the ubiquitin-proteasome system in monitoring and maintaining membrane lipid composition. We focus on the ER-resident RNF145 E3 ubiquitin ligase, its role in regulating adiponectin receptor 2 (ADIPOR2), its lipid hydrolase substrate, and the broader implications for understanding the homeostatic processes that fine-tune cellular membrane composition.
Collapse
Affiliation(s)
- Christian M Gawden-Bone
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Paul J Lehner
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Norbert Volkmar
- Institute for Molecular Systems Biology (IMSB), ETH Zürich, Zürich, Switzerland
| |
Collapse
|
4
|
Wan JX, Wang YQ, Lan SN, Chen L, Feng MQ, Chen X. Research Progress in Function and Regulation of E3 Ubiquitin Ligase SMURF1. Curr Med Sci 2023; 43:855-868. [PMID: 37558865 DOI: 10.1007/s11596-023-2774-x] [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: 05/18/2023] [Accepted: 06/08/2023] [Indexed: 08/11/2023]
Abstract
Smad ubiquitylation regulatory factor 1 (Smurf1) is an important homologous member of E6-AP C-terminus type E3 ubiquitin ligase. Initially, Smurf1 was reportedly involved in the negative regulation of the bone morphogenesis protein (BMP) pathway. After further research, several studies have confirmed that Smurf1 is widely involved in various biological processes, such as bone homeostasis regulation, cell migration, apoptosis, and planar cell polarity. At the same time, recent studies have provided a deeper understanding of the regulatory mechanisms of Smurf1's expression, activity, and substrate selectivity. In our review, a brief summary of recent important biological functions and regulatory mechanisms of E3 ubiquitin ligase Smurf1 is proposed.
Collapse
Affiliation(s)
- Ji-Xi Wan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yu-Qi Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Si-Na Lan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Liu Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ming-Qian Feng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xin Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
5
|
Niu H, Bi F, Zhao W, Xu Y, Han Q, Guo W, Chen Y. Smurf1 regulates ameloblast polarization by ubiquitination-mediated degradation of RhoA. Cell Prolif 2022; 56:e13387. [PMID: 36579844 PMCID: PMC10068949 DOI: 10.1111/cpr.13387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/30/2022] Open
Abstract
Cell polarity is essential for ameloblast differentiation and enamel formation. Smurf1 can mediate cell polarization through ubiquitination degradation of specific substrates. But it remains unclear whether Smurf1 could regulate ameloblast polarity and the underlying mechanism. Here, immuno-fluorescence staining and RT-qPCR were applied to detect the expression of Smurf1 and F-actin. A mouse lower incisor defect model was constructed. Scanning electron microscope, rat lower incisor culture, western blot, wound healing assay and trans-well migration assay were performed to detect the influence of Smurf1 knockdown on ameloblast. IF double staining, western blot and co-immunoprecipitation were conducted to detect the interaction between Smurf1 and RhoA. The in vivo experiment was also performed. We found that Smurf1 was mainly expressed in the membrane and cell cortex of ameloblast, similar to F-actin. Smurf1 expression increased along ameloblast polarization and differentiation. After knocking down Smurf1, the cytoskeleton and cell morphology changed and the cell polarity was damaged. Smurf1 regulated ameloblast polarity through ubiquitination degradation of activated RhoA in vitro. Local knockdown of Smurf1 in rat lower incisor ameloblast resulted in ameloblast polarity loss, enamel matrix secretion disorder and chalky enamel, but RhoA inhibitor Y-27632 could reverse this effect. Collectively, Smurf1 could regulate the polarization of ameloblast through ubiquitination degradation of activated RhoA, which contributed to the knowledge of tooth development and provided new research ideas for cell polarity.
Collapse
Affiliation(s)
- Haoman Niu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Fei Bi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wenjun Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yuchan Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qi Han
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Ravindran R, Velikkakath AKG, Narendradev ND, Chandrasekharan A, Santhoshkumar TR, Srinivasula SM. Endosomal-associated RFFL facilitates mitochondrial clearance by enhancing PRKN/parkin recruitment to mitochondria. Autophagy 2022; 18:2851-2864. [PMID: 35373701 PMCID: PMC9673925 DOI: 10.1080/15548627.2022.2052460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mutations in the ubiquitin ligase PRKN (parkin RBR E3 ubiquitin protein ligase) are associated with Parkinson disease and defective mitophagy. Conceptually, PRKN-dependent mitophagy is classified into two phases: 1. PRKN recruits to and ubiquitinates mitochondrial proteins; 2. formation of phagophore membrane, sequestering mitochondria for degradation. Recently, endosomal machineries are reported to contribute to the later stage for membrane assembly. We reported a role for endosomes in the events upstream of phase 1. We demonstrate that the endosomal ubiquitin ligase RFFL (ring finger and FYVE like domain containing E3 ubiquitin protein ligase) associated with damaged mitochondria, and this association preceded that of PRKN. RFFL interacted with PRKN, and stable recruitment of PRKN to damaged mitochondria was substantially reduced in RFFL KO cells. Our study unraveled a novel role of endosomes in modulating upstream pathways of PRKN-dependent mitophagy initiation.Abbreviations CCCP: carbonyl cyanide 3-chlorophenylhydrazone; DMSO: dimethyl sulfoxide; EGFP: enhanced green fluorescence protein; KO: knockout; PRKN: parkin RBR E3 ubiquitin protein ligase; RFFL: ring finger and FYVE like domain containing E3 ubiquitin protein ligase; UQCRC1: ubiquinol-cytochrome c reductase core protein 1; WT: wild-type.
Collapse
Affiliation(s)
- Rishith Ravindran
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| | - Anoop Kumar G. Velikkakath
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India,Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to Be University), Karnataka, India
| | - Nikhil Dev Narendradev
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| | | | - T. R. Santhoshkumar
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Kerala, India
| | - Srinivasa M. Srinivasula
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India,CONTACT Srinivasa M. Srinivasula School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala695551, India
| |
Collapse
|
7
|
Xu K, Chu Y, Liu Q, Fan W, He H, Huang F. NEDD4 E3 Ligases: Functions and Mechanisms in Bone and Tooth. Int J Mol Sci 2022; 23:ijms23179937. [PMID: 36077334 PMCID: PMC9455957 DOI: 10.3390/ijms23179937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022] Open
Abstract
Protein ubiquitination is a precisely controlled enzymatic cascade reaction belonging to the post-translational modification of proteins. In this process, E3 ligases catalyze the binding of ubiquitin (Ub) to protein substrates and define specificity. The neuronally expressed developmentally down-regulated 4 (NEDD4) subfamily, belonging to the homology to E6APC terminus (HECT) class of E3 ligases, has recently emerged as an essential determinant of multiple cellular processes in different tissues, including bone and tooth. Here, we place special emphasis on the regulatory role of the NEDD4 subfamily in the molecular and cell biology of osteogenesis. We elucidate in detail the specific roles, downstream substrates, and upstream regulatory mechanisms of the NEDD4 subfamily. Further, we provide an overview of the involvement of E3 ligases and deubiquitinases in the development, repair, and regeneration of another mineralized tissue—tooth.
Collapse
Affiliation(s)
- Ke Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Yanhao Chu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Qin Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Wenguo Fan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
- Correspondence: (H.H.); (F.H.)
| | - Fang Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
- Correspondence: (H.H.); (F.H.)
| |
Collapse
|
8
|
Tsunoda T, Riku M, Yamada N, Tsuchiya H, Tomita T, Suzuki M, Kizuki M, Inoko A, Ito H, Murotani K, Murakami H, Saeki Y, Kasai K. ENTREP/FAM189A2 encodes a new ITCH ubiquitin ligase activator that is downregulated in breast cancer. EMBO Rep 2022; 23:e51182. [PMID: 34927784 PMCID: PMC8811627 DOI: 10.15252/embr.202051182] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/13/2021] [Accepted: 11/29/2021] [Indexed: 11/09/2022] Open
Abstract
The HECT-type ubiquitin E3 ligases including ITCH regulate many aspects of cellular function through ubiquitinating various substrates. These ligases are known to be allosterically autoinhibited and to require an activator protein to fully achieve the ubiquitination of their substrates. Here we demonstrate that FAM189A2, a downregulated gene in breast cancer, encodes a new type of ITCH activator. FAM189A2 is a transmembrane protein harboring PPxY motifs, and the motifs mediate its association with and ubiquitination by ITCH. FAM189A2 also associates with Epsin and accumulates in early and late endosomes along with ITCH. Intriguingly, FAM189A2 facilitates the association of a chemokine receptor CXCR4 with ITCH and enhances ITCH-mediated ubiquitination of CXCR4. FAM189A2-knockout prohibits CXCL12-induced endocytosis of CXCR4, thereby enhancing the effects of CXCL12 on the chemotaxis and mammosphere formation of breast cancer cells. In comparison to other activators or adaptors known in the previous studies, FAM189A2 is a unique activator for ITCH to desensitize CXCR4 activity, and we here propose that FAM189A2 be renamed as ENdosomal TRansmembrane binding with EPsin (ENTREP).
Collapse
Affiliation(s)
- Takumi Tsunoda
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| | - Miho Riku
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| | - Norika Yamada
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| | - Hikaru Tsuchiya
- Protein Metabolism ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Takuya Tomita
- Protein Metabolism ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Minako Suzuki
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| | - Mari Kizuki
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| | - Akihito Inoko
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
| | - Hideaki Ito
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| | | | - Hideki Murakami
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| | - Yasushi Saeki
- Protein Metabolism ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Kenji Kasai
- Department of PathologyAichi Medical University School of MedicineNagakuteJapan
| |
Collapse
|
9
|
Han D, Li S, Xia Q, Meng X, Dong L. Overexpressed Smurf1 is degraded in glioblastoma cells through autophagy in a p62-dependent manner. FEBS Open Bio 2022; 12:118-129. [PMID: 34614303 PMCID: PMC8727935 DOI: 10.1002/2211-5463.13310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 11/07/2022] Open
Abstract
Homologous to E6AP C-terminus (HECT)-type E3 ubiquitin ligase SMAD-specific E3 ubiquitin protein ligase 1 (Smurf1) was originally identified to ubiquitinate Smad protein in the TGF-β/BMP signaling pathway. Recently, Smurf1 has been reported to promote tumorigenesis by regulating multiple biological processes. High expression of Smurf1 plays a vital role in brain tumor progression by mediating aberrant cell signaling pathways. Previous reports have shown that Smurf1 is degraded mainly through the ubiquitin-proteasome system, but it remains unclear whether Smurf1 is degraded by autophagy in tumor cells. In this study, we show that autophagy activators promote Smurf1 degradation in glioblastoma (GB) cells. The autophagy receptor p62 colocalizes with ubiquitinated substrates to promote sequestration of cytoplasm cargo into the autophagosome. We report that autophagic degradation of Smurf1 is dependent on p62. Moreover, the autophagic degradation of Smurf1 is prevented in the absence of the HECT domain or E3 ubiquitin ligase activity. We further proved that activation of autophagy leads to a decrease of Smurf1 and the inhibition of the phosphoinositide 3-kinase/protein kinase B signaling pathway in GB cells. Our results suggest that enhancement of autophagic degradation of Smurf1 may be a potential approach to treating GB.
Collapse
Affiliation(s)
- Da Han
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Shengzhen Li
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Qin Xia
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Xinyi Meng
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Lei Dong
- School of Life ScienceBeijing Institute of TechnologyChina
| |
Collapse
|
10
|
Li Y, Cui C, Xie F, Kiełbasa S, Mei H, van Dinther M, van Dam H, Bauer A, Zhang L, Ten Dijke P. VprBP mitigates TGF-β and Activin signaling by promoting Smurf1-mediated type I receptor degradation. J Mol Cell Biol 2021; 12:138-151. [PMID: 31291647 PMCID: PMC7109606 DOI: 10.1093/jmcb/mjz057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/16/2019] [Accepted: 06/06/2019] [Indexed: 12/13/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family controls embryogenesis, stem cell differentiation, and tissue homeostasis. However, how post-translation modifications contribute to fine-tuning of TGF-β family signaling responses is not well understood. Inhibitory (I)-Smads can antagonize TGF-β/Smad signaling by recruiting Smurf E3 ubiquitin ligases to target the active TGF-β receptor for proteasomal degradation. A proteomic interaction screen identified Vpr binding protein (VprBP) as novel binding partner of Smad7. Mis-expression studies revealed that VprBP negatively controls Smad2 phosphorylation, Smad2-Smad4 interaction, as well as TGF-β target gene expression. VprBP was found to promote Smad7-Smurf1-TβRI complex formation and induce proteasomal degradation of TGF-β type I receptor (TβRI). Moreover, VprBP appears to stabilize Smurf1 by suppressing Smurf1 poly-ubiquitination. In multiple adult and mouse embryonic stem cells, depletion of VprBP promotes TGF-β or Activin-induced responses. In the mouse embryo VprBP expression negatively correlates with mesoderm marker expression, and VprBP attenuated mesoderm induction during zebrafish embryogenesis. Our findings thereby uncover a novel regulatory mechanism by which Smurf1 controls the TGF-β and Activin cascade and identify VprBP as a critical determinant of embryonic mesoderm induction.
Collapse
Affiliation(s)
- Yihao Li
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Chao Cui
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Feng Xie
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Szymon Kiełbasa
- Department of Human Genetics, Leiden Genome Technology Centre, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Hailiang Mei
- Sequence Analysis Support Core, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Maarten van Dinther
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Hans van Dam
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Andreas Bauer
- Novartis Institutes for BioMedical Research, Inc., Novartis Campus, Forum 2.5.01.30, CH-4056, Basel, Switzerland
| | - Long Zhang
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands.,MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands.,MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
11
|
Xia Q, Li Y, Han D, Dong L. SMURF1, a promoter of tumor cell progression? Cancer Gene Ther 2020; 28:551-565. [PMID: 33204002 DOI: 10.1038/s41417-020-00255-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022]
Abstract
Overexpression of HECT-type E3 ubiquitin ligase SMURF1 is correlated with poor prognosis in patients with various cancers, such as glioblastoma, colon cancer, and clear cell renal cell carcinoma. SMURF1 acts as a tumor promoter by ubiquitination modification and/or degradation of tumor-suppressing proteins. Combined treatment of Smurf1 knockdown with rapamycin showed collaborative antitumor effects in mice. This review described the role of HECT, WW, and C2 domains in regulating SMURF1 substrate selection. We summarized up to date SMURF1 substrates regulating different type cell signaling, thus, accelerating tumor progression, invasion, and metastasis. Furthermore, the downregulation of SMURF1 expression, inhibition of its E3 activity and regulation of its specificity to substrates prevent tumor progression. The potential application of SMURF1 regulators, specifically, wisely choose certain drugs by blocking SMURF1 selectivity in tumor suppressors, to develop novel anticancer treatments.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Da Han
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
12
|
Scott JL, Frick CT, Johnson KA, Liu H, Yong SS, Varney AG, Wiest O, Stahelin RV. Molecular Analysis of Membrane Targeting by the C2 Domain of the E3 Ubiquitin Ligase Smurf1. Biomolecules 2020; 10:biom10020229. [PMID: 32033048 PMCID: PMC7072158 DOI: 10.3390/biom10020229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
SMAD ubiquitination regulatory factor 1 (Smurf1) is a Nedd4 family E3 ubiquitin ligase that regulates cell motility, polarity and TGFβ signaling. Smurf1 contains an N-terminal protein kinase C conserved 2 (C2) domain that targets cell membranes and is required for interactions with membrane-localized substrates such as RhoA. Here, we investigated the lipid-binding mechanism of Smurf1 C2, revealing a general affinity for anionic membranes in addition to a selective affinity for phosphoinositides (PIPs). We found that Smurf1 C2 localizes not only to the plasma membrane but also to negatively charged intracellular sites, acting as an anionic charge sensor and selective PIP-binding domain. Site-directed mutagenesis combined with docking/molecular dynamics simulations revealed that the Smurf1 C2 domain loop region primarily interacts with PIPs and cell membranes, as opposed to the β-surface cationic patch employed by other C2 domains. By depleting PIPs from the inner leaflet of the plasma membrane, we found that PIP binding is necessary for plasma membrane localization. Finally, we used a Smurf1 cellular ubiquitination assay to show that the amount of ubiquitin at the plasma membrane interface depends on the lipid-binding properties of Smurf1. This study shows the mechanism by which Smurf1 C2 targets membrane-based substrates and reveals a novel interaction for non-calcium-dependent C2 domains and membrane lipids.
Collapse
Affiliation(s)
- Jordan L. Scott
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Cary T. Frick
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Kristen A. Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Haining Liu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Sylvia S. Yong
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Allyson G. Varney
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Olaf Wiest
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Robert V. Stahelin
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: ; Tel.: +1-765-494-4152
| |
Collapse
|
13
|
Fu L, Cui CP, Zhang X, Zhang L. The functions and regulation of Smurfs in cancers. Semin Cancer Biol 2019; 67:102-116. [PMID: 31899247 DOI: 10.1016/j.semcancer.2019.12.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/10/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023]
Abstract
Smad ubiquitination regulatory factor 1 (Smurf1) and Smurf2 are HECT-type E3 ubiquitin ligases, and both Smurfs were initially identified to regulate Smad protein stability in the TGF-β/BMP signaling pathway. In recent years, Smurfs have exhibited E3 ligase-dependent and -independent activities in various kinds of cells. Smurfs act as either potent tumor promoters or tumor suppressors in different tumors by regulating biological processes, including metastasis, apoptosis, cell cycle, senescence and genomic stability. The regulation of Smurfs activity and expression has therefore emerged as a hot spot in tumor biology research. Further, the Smurf1- or Smurf2-deficient mice provide more in vivo clues for the functional study of Smurfs in tumorigenesis and development. In this review, we summarize these milestone findings and, in turn, reveal new avenues for the prevention and treatment of cancer by regulating Smurfs.
Collapse
Affiliation(s)
- Lin Fu
- Institute of Chronic Disease, Qingdao Municipal Hospital, Qingdao University, Qingdao 266000, China
| | - Chun-Ping Cui
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| | - Xueli Zhang
- Department of General Surgery, Shanghai Fengxian Central Hospital Graduate Training Base, Fengxian Hospital, Southern Medical University, Shanghai, China.
| | - Lingqiang Zhang
- Institute of Chronic Disease, Qingdao Municipal Hospital, Qingdao University, Qingdao 266000, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; Peixian People's Hospital, Jiangsu Province 221600, China.
| |
Collapse
|
14
|
Ruetalo N, Anders S, Stollmaier C, Jäckl M, Schütz-Stoffregen MC, Stefan N, Wolf C, Wiesner S. The WW1 Domain Enhances Autoinhibition in Smurf Ubiquitin Ligases. J Mol Biol 2019; 431:4834-4847. [PMID: 31628949 DOI: 10.1016/j.jmb.2019.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
Abstract
Downregulation of ubiquitin (Ub) ligase activity prevents premature ubiquitination and is critical for cellular homeostasis. Nedd4 Ub ligases share a common domain architecture and yet are regulated in distinct ways through interactions of the catalytic HECT domain with the N-terminal C2 domain or the central WW domain region. Smurf1 and Smurf2 are two highly related Nedd4 ligases with ~70% overall sequence identity. Here, we show that the Smurf1 C2 domain interacts with the HECT domain and inhibits ligase activity in trans. However, in contrast to Smurf2, we find that full-length Smurf1 is a highly active Ub ligase, and we can attribute this striking difference in regulation to the lack of one WW domain (WW1) in Smurf1. Using NMR spectroscopy and biochemical assays, we identified the WW1 region as an additional inhibitory element in Smurf2 that cooperates with the C2 domain to enhance HECT domain binding and Smurf2 inhibition. Our work provides important insights into Smurf regulation and highlights that the activities of highly related proteins can be controlled in distinct ways.
Collapse
Affiliation(s)
- Natalia Ruetalo
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Samira Anders
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Carsten Stollmaier
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany; Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Magnus Jäckl
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Mira C Schütz-Stoffregen
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany; Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Nadine Stefan
- Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Christine Wolf
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Silke Wiesner
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany; Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany.
| |
Collapse
|
15
|
Jiang M, Shi L, Yang C, Ge Y, Lin L, Fan H, He Y, Zhang D, Miao Y, Yang L. miR-1254 inhibits cell proliferation, migration, and invasion by down-regulating Smurf1 in gastric cancer. Cell Death Dis 2019; 10:32. [PMID: 30631050 PMCID: PMC6328618 DOI: 10.1038/s41419-018-1262-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/05/2018] [Accepted: 12/03/2018] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is one of the most frequent malignancies, and increasing evidence supports the contribution of microRNA (miRNAs) to cancer progression. miR-1254 has been confirmed to participate in the regulation of various cancers, while the function of miR-1254 in GC remains unknown. In this study, we investigated the role of miR-1254 in GC. The expression of miR-1254 was detected in human GC specimens and cell lines by miRNA RT-PCR. The effects of miR-1254 on GC proliferation were determined by CCK-8 proliferation assays, colony formation assays, 5-ethynyl-2'-deoxyuridine (EdU) incorporation, and cell-cycle assays. The ability of migration and invasion was examined by transwell and wound-healing assay. Dual Luciferase reporter assay was used to validate the interaction of miR-1254 with its target gene. The xenograft mouse models were conducted to investigate the effects of miR-1254 in vivo. The signaling pathways and epithelial-mesenchymal transition (EMT)-related proteins were detected with western blot. The results showed that miR-1254 inhibited the proliferation, migration and invasion in vitro and suppressed tumorigenesis in vivo. Smurf1 was shown to be the direct target of miR-1254. Overexpressing Smurf1 could partially counteract the effects caused by miR-1254. Similarly, the effects of the miR-1254-inhibitor were also rescued by Smurf1-shRNA. Furthermore, we found that miR-1254 inhibited EMT and decreased the PI3K/AKT signaling pathway through downregulating Smurf1. In summary, overexpression of miR-1254 could suppress proliferation, migration, invasion, and EMT via PI3K/AKT signaling pathways by downregulation of Smurf1 in GC, which suggests a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Mingkun Jiang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Shi
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chao Yang
- Department of Liver Surgery/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission of China, Nanjing, China
| | - Yugang Ge
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Linling Lin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Fan
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu He
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongchang Miao
- Department of General Surgery, the second People's Hospital of Lianyungang, Lianyungang, China.
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
16
|
Werner CT, Viswanathan R, Martin JA, Gobira PH, Mitra S, Thomas SA, Wang ZJ, Liu JF, Stewart AF, Neve RL, Li JX, Gancarz AM, Dietz DM. E3 Ubiquitin-Protein Ligase SMURF1 in the Nucleus Accumbens Mediates Cocaine Seeking. Biol Psychiatry 2018; 84:881-892. [PMID: 30158054 PMCID: PMC6260585 DOI: 10.1016/j.biopsych.2018.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/25/2018] [Accepted: 07/10/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Substance use disorder is a neurobiological disease characterized by episodes of relapse despite periods of withdrawal. It is thought that neuroadaptations in discrete brain areas of the reward pathway, including the nucleus accumbens, underlie these aberrant behaviors. The ubiquitin-proteasome system degrades proteins and has been shown to be involved in cocaine-induced plasticity, but the role of E3 ubiquitin ligases, which conjugate ubiquitin to substrates, is unknown. Here, we examined E3 ubiquitin-protein ligase SMURF1 (SMURF1) in neuroadaptations and relapse behavior during withdrawal following cocaine self-administration. METHODS SMURF1 and downstream targets ras homolog gene family, member A (RhoA), SMAD1/5, and Runt-related transcript factor 2 were examined using Western blotting (n = 9-11/group), quantitative polymerase chain reaction (n = 6-9/group), co-immunoprecipitation (n = 9-11/group), tandem ubiquitin binding entities affinity purification (n = 5-6/group), and quantitative chromatin immunoprecipitation (n = 3-6/group) (2 rats/sample). Viral-mediated gene transfer (n = 7-12/group) and intra-accumbal microinjections (n = 9-10/group) were used to examine causal roles of SMURF1 and substrate RhoA, respectively, in cue-induced cocaine seeking. RESULTS SMURF1 protein expression was decreased, while SMURF1 substrates RhoA and SMAD1/5 were increased, in the nucleus accumbens on withdrawal day 7, but not on withdrawal day 1, following cocaine self-administration. Viral-mediated gene transfer of Smurf1 or constitutive activation of RhoA attenuated cue-induced cocaine seeking, while catalytically inactive Smurf1 enhanced cocaine seeking. Furthermore, SMURF1-regulated, SMAD1/5-associated transcription factor Runt-related transcript factor 2 displayed increased binding at promoter regions of genes previously associated with cocaine-induced plasticity. CONCLUSIONS SMURF1 is a key mediator of neuroadaptations in the nucleus accumbens following cocaine exposure and mediates cue-induced cocaine seeking during withdrawal.
Collapse
Affiliation(s)
- Craig T Werner
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Rathipriya Viswanathan
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Jennifer A Martin
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Pedro H Gobira
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York; Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Swarup Mitra
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Shruthi A Thomas
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Zi-Jun Wang
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Jian-Feng Liu
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Andrew F Stewart
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Cambridge, Massachusetts
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Amy M Gancarz
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York; Department of Psychology, California State University, Bakersfield, Bakersfield, California
| | - David M Dietz
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York; Department of Psychology, The State University of New York at Buffalo, Buffalo, New York.
| |
Collapse
|
17
|
Murakami K, Etlinger JD. Role of SMURF1 ubiquitin ligase in BMP receptor trafficking and signaling. Cell Signal 2018; 54:139-149. [PMID: 30395943 DOI: 10.1016/j.cellsig.2018.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 11/28/2022]
Abstract
Heterozygous germline mutations in the bone morphogenetic protein type II receptor gene (BMPRII) are associated with hereditary pulmonary arterial hypertension (HPAH). Missense mutations, both in the extracellular ligand-binding and cytoplasmic kinase domains, mostly involve substitution of conserved Cys residues. Singular substitution at any of those Cys residues causes cytoplasmic, perinuclear localization of BMPR with reduced cell surface expression and BMP signaling. The present study examined the effect of Cys residue substitution on BMPR endocytic trafficking and lysosome degradation. We demonstrate that endocytosis/lysosomal degradation of BMPR occurs by two distinct pathways. SMURF1 ubiquitin ligase induces lysosomal degradation of BMPR, while ligase-inactive SMURF1 maintains BMPR protein level and cell surface expression. Substitution of BMPR Cys residues increases lysosomal degradation which is blocked by ligase-inactive SMURF1, elevating protein levels of Cys-substituted BMPRs. Expression of Cys-substituted BMPR suppresses basal BMP signaling activity which is also up-regulated by ligase-inactive SMURF1. Cys-residue substitution thus appears to cause BMPR endocytosis to lysosomes in a SMURF1 ubiquitin ligase-associated pathway. Kinase-activated BMPR undergoes endocytic/lysosomal degradation by a pathway with certain unique properties. Therefore, our results describe a novel mechanism whereby SMURF1 ubiquitin ligase regulates constitutive endocytosis of BMPR which may be mediated by its conserved Cys residues.
Collapse
Affiliation(s)
- Koko Murakami
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595, USA.
| | - Joseph D Etlinger
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595, USA
| |
Collapse
|
18
|
Todaro DR, Augustus-Wallace AC, Klein JM, Haas AL. The mechanism of neural precursor cell expressed developmentally down-regulated 4-2 (Nedd4-2)/NEDD4L-catalyzed polyubiquitin chain assembly. J Biol Chem 2017; 292:19521-19536. [PMID: 28972136 DOI: 10.1074/jbc.m117.817882] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Indexed: 11/06/2022] Open
Abstract
The mechanism of Nedd4-2 has been quantitatively explored for the first time using biochemically defined kinetic assays examining rates of 125I-polyubiquitin chain assembly as a functional readout. We demonstrate that Nedd4-2 exhibits broad specificity for E2 paralogs of the Ubc4/5 clade to assemble Lys63-linked polyubiquitin chains. Full-length Nedd4-2 catalyzes free 125I-polyubiquitin chain assembly by hyperbolic Michaelis-Menten kinetics with respect to Ubc5B∼ubiquitin thioester concentration (Km = 44 ± 6 nm; kcat = 0.020 ± 0.007 s-1) and substrate inhibition above 0.5 μm (Ki = 2.5 ± 1.3 μm) that tends to zero velocity, requiring ordered binding at two functionally distinct E2∼ubiquitin-binding sites. The Ubc5BC85A product analog non-competitively inhibits Nedd4-2 (Ki = 2.0 ± 0.5 μm), consistent with the presence of the second E2-binding site. In contrast, the isosteric Ubc5BC85S-ubiquitin oxyester substrate analog exhibits competitive inhibition at the high-affinity Site 1 (Ki = 720 ± 340 nm) and non-essential activation at the lower-affinity Site 2 (Kact = 750 ± 260 nm). Additional studies utilizing Ubc5BF62A, defective in binding the canonical E2 site, demonstrate that the cryptic Site 1 is associated with thioester formation, whereas binding at the canonical site (Site 2) is associated with polyubiquitin chain elongation. Finally, previously described Ca2+-dependent C2 domain-mediated autoinhibition of Nedd4-2 is not observed under our reported experimental conditions. These studies collectively demonstrate that Nedd4-2 catalyzes polyubiquitin chain assembly by an ordered two-step mechanism requiring two dynamically linked E2∼ubiquitin-binding sites analogous to that recently reported for E6AP, the founding member of the Hect ligase family.
Collapse
Affiliation(s)
- Dustin R Todaro
- From the Department of Biochemistry and Molecular Biology and
| | | | | | - Arthur L Haas
- From the Department of Biochemistry and Molecular Biology and .,the Stanley S. Scott Cancer Center, Louisiana State University School of Medicine, New Orleans, Louisiana 70112
| |
Collapse
|
19
|
Smurf1 regulates lung cancer cell growth and migration through interaction with and ubiquitination of PIPKIγ. Oncogene 2017; 36:5668-5680. [DOI: 10.1038/onc.2017.166] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/31/2017] [Accepted: 04/26/2017] [Indexed: 12/12/2022]
|
20
|
Franco LH, Nair VR, Scharn CR, Xavier RJ, Torrealba JR, Shiloh MU, Levine B. The Ubiquitin Ligase Smurf1 Functions in Selective Autophagy of Mycobacterium tuberculosis and Anti-tuberculous Host Defense. Cell Host Microbe 2017; 21:59-72. [PMID: 28017659 PMCID: PMC5699477 DOI: 10.1016/j.chom.2016.11.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 10/14/2016] [Accepted: 11/18/2016] [Indexed: 01/03/2023]
Abstract
During antibacterial autophagy, ubiquitination of intracellular bacteria recruits proteins that mediate bacterial delivery to the lysosome for degradation. Smurf1 is an E3 ubiquitin ligase whose role in selective bacterial autophagy is unknown. We show that Smurf1 facilitates selective autophagy of the human pathogen Mycobacterium tuberculosis (Mtb). Smurf1-/- macrophages are defective in recruiting polyubiquitin, the proteasome, the ubiquitin-binding autophagy adaptor NBR1, the autophagy protein LC3, and the lysosomal marker LAMP1 to Mtb-associated structures and are more permissive for Mtb growth. This function of Smurf1 requires both its ubiquitin-ligase and C2 phospholipid-binding domains, and involves K48- rather than K63-linked ubiquitination. Chronically infected Smurf1-/- mice have increased bacterial load, increased lung inflammation, and accelerated mortality. SMURF1 controls Mtb replication in human macrophages and associates with bacteria in lungs of patients with pulmonary tuberculosis. Thus, Smurf1 is required for selective autophagy of Mtb and host defense against tuberculosis infection.
Collapse
Affiliation(s)
- Luis H Franco
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vidhya R Nair
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Caitlyn R Scharn
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ramnik J Xavier
- Gastrointestinal Unit and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02142, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Jose R Torrealba
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael U Shiloh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Beth Levine
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
21
|
Bouin AP, Kyurmurkov A, Régent-Kloeckner M, Ribba AS, Faurobert E, Fournier HN, Bourrin-Reynard I, Manet-Dupé S, Oddou C, Balland M, Planus E, Albiges-Rizo C. ICAP-1 monoubiquitination coordinates matrix density and rigidity sensing for cell migration through ROCK2- MRCKα balance. J Cell Sci 2017; 130:626-636. [DOI: 10.1242/jcs.200139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 12/08/2016] [Indexed: 12/21/2022] Open
Abstract
Cell migration is a complex process requiring density and rigidity sensing of the microenvironment to adapt cell migratory speed through focal adhesion and actin cytoskeleton regulation. ICAP-1, a β1 integrin partner, is essential for ensuring integrin activation cycle and focal adhesion formation. We show that ICAP-1 is monoubiquitinated by Smurf1, preventing ICAP-1 binding to β1 integrin. The non-ubiquitinable form of ICAP-1 modifies β1 integrin focal adhesion organization and interferes with fibronectin density sensing. ICAP-1 is also required for adapting cell migration in response to substrate stiffness in a β1 integrin-independent manner. ICAP-1 monoubiquitination regulates rigidity sensing by increasing MRCKα-dependent cell contractility through myosin phosphorylation independently of substrate rigidity. We provide evidence that ICAP-1 monoubiquitination helps in switching from ROCK2-mediated to MRCKα-mediated cell contractility. ICAP-1 monoubiquitination serves as a molecular switch to coordinate extracellular matrix density and rigidity sensing thus acting as a critical modulator of cell migration and mechanosensing.
Collapse
Affiliation(s)
- Anne-Pascale Bouin
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Alexander Kyurmurkov
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Myriam Régent-Kloeckner
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Anne-Sophie Ribba
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Eva Faurobert
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Henri-Noël Fournier
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Ingrid Bourrin-Reynard
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Sandra Manet-Dupé
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Christiane Oddou
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Martial Balland
- CNRS UMR 5309, F-38042 Grenoble, France
- Laboratoire Interdisciplinaire de Physique, UMR CNRS 5588Grenoble, France
| | - Emmanuelle Planus
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| | - Corinne Albiges-Rizo
- INSERM U1209, Grenoble, F-38042, France
- Université Grenoble Alpes, Institute for Advanced Biosciences, 38042 Grenoble, France
- CNRS UMR 5309, F-38042 Grenoble, France
| |
Collapse
|
22
|
Abstract
The virulence of highly pathogenic bacteria such as Salmonella, Yersinia, Staphylococci, Clostridia, and pathogenic strains of Escherichia coli involves intimate cross-talks with the host actin cytoskeleton and its upstream regulators. A large number of virulence factors expressed by these pathogens modulate Rho GTPase activities either by mimicking cellular regulators or by catalyzing posttranslational modifications of these small proteins. This impressive convergence of virulence toward Rho GTPases and actin indeed offers pathogens the capacity to breach host defenses and invade their host, while it promotes inflammatory reactions. In return, the study of this targeting of Rho GTPases in infection has been an invaluable source of information in cell signaling, cell biology, and biomechanics, as well as in immunology. Through selected examples, I highlight the importance of recent studies on this crosstalk, which have unveiled new mechanisms of regulation of Rho GTPases; the relationship between cell shape and actin cytoskeleton organization; and the relationship between Rho GTPases and innate immune signaling.
Collapse
Affiliation(s)
- Emmanuel Lemichez
- UCA, Inserm, C3M, U1065, Team Microbial Toxins in Host Pathogen Interactions, Equipe Labellisée la Ligue Contre le Cancer, Nice, 06204, France.
- UFR Médecine, Université de Nice-Sophia-Antipolis, Nice, France.
| |
Collapse
|
23
|
Liu ZQ, Qiu AL, Shi LP, Cai JS, Huang XY, Yang S, Wang B, Shen L, Huang MK, Mou SL, Ma XL, Liu YY, Lin L, Wen JY, Tang Q, Shi W, Guan DY, Lai Y, He SL. SRC2-1 is required in PcINF1-induced pepper immunity by acting as an interacting partner of PcINF1. JOURNAL OF EXPERIMENTAL BOTANY 2015; 66:3683-98. [PMID: 25922484 DOI: 10.1093/jxb/erv161] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Elicitins are elicitors that can trigger hypersensitive cell death in most Nicotiana spp., but their underlying molecular mechanism is not well understood. The gene Phytophthora capsici INF1 (PcINF1) coding for an elicitin from P. capsici was characterized in this study. Transient overexpression of PcINF1 triggered cell death in pepper (Capsicum annuum L.) and was accompanied by upregulation of the hypersensitive response marker, Hypersensitive Induced Reaction gene 1 (HIR1), and the pathogenesis-related genes SAR82, DEF1, BPR1, and PO2. A putative PcINF1-interacting protein, SRC2-1, was isolated from a pepper cDNA library by yeast two-hybrid screening and was observed to target the plasma membrane. The interaction between PcINF1 and SRC2-1 was confirmed by bimolecular fluorescence complementation and co-immunoprecipitation. Simultaneous transient overexpression of SRC2-1 and PcINF1 in pepper plants triggered intensive cell death, whereas silencing of SRC2-1 by virus-induced gene silencing blocked the cell death induction of PcINF1 and increased the susceptibility of pepper plants to P. capsici infection. Additionally, membrane targeting of the PcINF1-SRC2-1 complex was required for cell death induction. The C2 domain of SRC2-1 was crucial for SRC2-1 plasma membrane targeting and the PcINF1-SRC2-1 interaction. These results suggest that SRC2-1 interacts with PcINF1 and is required in PcINF1-induced pepper immunity.
Collapse
Affiliation(s)
- Zhi-qin Liu
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Ai-lian Qiu
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Lan-ping Shi
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Jin-sen Cai
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Xue-ying Huang
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Sheng Yang
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Bo Wang
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Lei Shen
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Mu-kun Huang
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Shao-liang Mou
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Xiao-Ling Ma
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Yan-yan Liu
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Lin Lin
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Jia-yu Wen
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Qian Tang
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Wei Shi
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - De-yi Guan
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Yan Lai
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| | - Shui-lin He
- National Education Minster Key Laboratory of Plant Genetic Improvement and Comprehensive Utilization, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China
| |
Collapse
|
24
|
Expression and purification of human WWP2 HECT domain in Escherichia coli. Protein Expr Purif 2015; 110:95-101. [DOI: 10.1016/j.pep.2014.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/10/2014] [Accepted: 12/19/2014] [Indexed: 11/23/2022]
|
25
|
Jiang J, Wang N, Jiang Y, Tan H, Zheng J, Chen G, Jia Z. Characterization of substrate binding of the WW domains in human WWP2 protein. FEBS Lett 2015; 589:1935-42. [PMID: 25999310 DOI: 10.1016/j.febslet.2015.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/26/2015] [Accepted: 05/05/2015] [Indexed: 10/23/2022]
Abstract
WW domains harbor substrates containing proline-rich motifs, but the substrate specificity and binding mechanism remain elusive for those WW domains less amenable for structural studies, such as human WWP2 (hWWP2). Herein we have employed multiple techniques to investigate the second WW domain (WW2) in hWWP2. Our results show that hWWP2 is a specialized E3 for PPxY motif-containing substrates only and does not recognize other amino acids and phospho-residues. The strongest binding affinity of WW2, and the incompatibility between each WW domain, imply a novel relationship, and our SPR experiment reveals a dynamic binding mode in Class-I WW domains for the first time. The results from alanine-scanning mutagenesis and modeling further point to functionally conserved residues in WW2.
Collapse
Affiliation(s)
- Jiahong Jiang
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Nan Wang
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Yafei Jiang
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Hongwei Tan
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Jimin Zheng
- College of Chemistry, Beijing Normal University, Beijing, China.
| | - Guangju Chen
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Zongchao Jia
- Department of Biochemical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
26
|
Structural and Functional Framework for the Autoinhibition of Nedd4-Family Ubiquitin Ligases. Structure 2014; 22:1639-49. [DOI: 10.1016/j.str.2014.09.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 09/03/2014] [Accepted: 09/10/2014] [Indexed: 01/22/2023]
|
27
|
Wang M, Guo L, Wu Q, Zeng T, Lin Q, Qiao Y, Wang Q, Liu M, Zhang X, Ren L, Zhang S, Pei Y, Yin Z, Ding F, Wang HR. ATR/Chk1/Smurf1 pathway determines cell fate after DNA damage by controlling RhoB abundance. Nat Commun 2014; 5:4901. [PMID: 25249323 DOI: 10.1038/ncomms5901] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 08/02/2014] [Indexed: 11/09/2022] Open
Abstract
ATM- and RAD3-related (ATR)/Chk1 and ataxia-telangiectasia mutated (ATM)/Chk2 signalling pathways play critical roles in the DNA damage response. Here we report that the E3 ubiquitin ligase Smurf1 determines cell apoptosis rates downstream of DNA damage-induced ATR/Chk1 signalling by promoting degradation of RhoB, a small GTPase recognized as tumour suppressor by promoting death of transformed cells. We show that Smurf1 targets RhoB for degradation to control its abundance in the basal state. DNA damage caused by ultraviolet light or the alkylating agent methyl methanesulphonate strongly activates Chk1, leading to phosphorylation of Smurf1 that enhances its self-degradation, hence resulting in a RhoB accumulation to promote apoptosis. Suppressing RhoB levels by overexpressing Smurf1 or blocking Chk1-dependent Smurf1 self-degradation significantly inhibits apoptosis. Hence, our study unravels a novel ATR/Chk1/Smurf1/RhoB pathway that determines cell fate after DNA damage, and raises the possibility that aberrant upregulation of Smurf1 promotes tumorigenesis by excessively targeting RhoB for degradation.
Collapse
Affiliation(s)
- Meilin Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lei Guo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Qingang Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Taoling Zeng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Qi Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yikai Qiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Qun Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Mingdong Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xin Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lan Ren
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Sheng Zhang
- Department of Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361005, China
| | - Yihua Pei
- Central Laboratory, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhenyu Yin
- Department of Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361005, China
| | - Feng Ding
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Hong-Rui Wang
- 1] State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China [2] Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong 518057, China
| |
Collapse
|
28
|
Cao Y, Wang C, Zhang X, Xing G, Lu K, Gu Y, He F, Zhang L. Selective small molecule compounds increase BMP-2 responsiveness by inhibiting Smurf1-mediated Smad1/5 degradation. Sci Rep 2014; 4:4965. [PMID: 24828823 PMCID: PMC4021816 DOI: 10.1038/srep04965] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 04/25/2014] [Indexed: 12/21/2022] Open
Abstract
The ubiquitin ligase Smad ubiquitination regulatory factor-1 (Smurf1) negatively regulates bone morphogenetic protein (BMP) pathway by ubiquitinating certain signal components for degradation. Thus, it can be an eligible pharmacological target for increasing BMP signal responsiveness. We established a strategy to discover small molecule compounds that block the WW1 domain of Smurf1 from interacting with Smad1/5 by structure based virtual screening, molecular experimental examination and cytological efficacy evaluation. Our selected hits could reserve the protein level of Smad1/5 from degradation by interrupting Smurf1-Smad1/5 interaction and inhibiting Smurf1 mediated ubiquitination of Smad1/5. Further, these compounds increased BMP-2 signal responsiveness and the expression of certain downstream genes, enhanced the osteoblastic activity of myoblasts and osteoblasts. Our work indicates targeting Smurf1 for inhibition could be an accessible strategy to discover BMP-sensitizers that might be applied in future clinical treatments of bone disorders such as osteopenia.
Collapse
Affiliation(s)
- Yu Cao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Cheng Wang
- 1] State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China [2] School of Medicine, Shihezi University, Shihezi, Xinjiang Province, China
| | - Xueli Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Guichun Xing
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Kefeng Lu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Yongqing Gu
- School of Medicine, Shihezi University, Shihezi, Xinjiang Province, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Lingqiang Zhang
- 1] State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China [2] Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
29
|
Fei C, He X, Xie S, Miao H, Zhou Z, Li L. Smurf1-mediated axin ubiquitination requires Smurf1 C2 domain and is cell cycle-dependent. J Biol Chem 2014; 289:14170-7. [PMID: 24700460 DOI: 10.1074/jbc.m113.536714] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously, Smad ubiquitination regulatory factor 1 (Smurf1)-mediated Lys29 (K29)-linked poly-ubiquitination of Axin has been identified as a novel regulatory process in Wnt/β-catenin signaling. In this work, we discovered that the C2 domain of Smurf1 is critical for targeting Axin for ubiquitination. We found that the C2 domain-mediated plasma membrane localization of Smurf1 is required for Axin ubiquitination, and interfering with that disturbs the co-localization of Smurf1 and Axin around the plasma membrane. Moreover, the C2 domain of Smurf1, rather than its WW domains, is involved in Smurf1's interaction with Axin; and the putative PPXY motifs (PY motif) of Axin are not essential for such an interaction, indicating that Smurf1 binds to Axin in a non-canonical way independent of WW-PY interaction. Further, we found that Smurf1-Axin interaction and Axin ubiquitination are attenuated in the G2/M phase of cell cycle, contributing to an increased cell response to Wnt stimulation at that stage. Collectively, we uncovered a dual role of Smurf1 C2 domain, recruiting Smurf1 to membrane for accessing Axin and mediating its interaction with Axin, and that Smurf1-mediated Axin ubiquitination is subjected to the regulation of cell cycle.
Collapse
Affiliation(s)
- Cong Fei
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoli He
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sichun Xie
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haofei Miao
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhilei Zhou
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lin Li
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
30
|
Palikaras K, Tavernarakis N. Mitochondrial homeostasis: the interplay between mitophagy and mitochondrial biogenesis. Exp Gerontol 2014; 56:182-8. [PMID: 24486129 DOI: 10.1016/j.exger.2014.01.021] [Citation(s) in RCA: 326] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/15/2014] [Accepted: 01/20/2014] [Indexed: 12/29/2022]
Abstract
Mitochondria are highly dynamic organelles and their proper function is crucial for the maintenance of cellular homeostasis. Mitochondrial biogenesis and mitophagy are two pathways that regulate mitochondrial content and metabolism preserving homeostasis. The tight regulation between these opposing processes is essential for cellular adaptation in response to cellular metabolic state, stress and other intracellular or environmental signals. Interestingly, imbalance between mitochondrial proliferation and degradation process results in progressive development of numerous pathologic conditions. Here we review recent studies that highlight the intricate interplay between mitochondrial biogenesis and mitophagy, mainly focusing on the molecular mechanisms that govern the coordination of these processes and their involvement in age-related pathologies and ageing.
Collapse
Affiliation(s)
- Konstantinos Palikaras
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 71110, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 71110, Crete, Greece.
| |
Collapse
|
31
|
Cao Y, Zhang L. A Smurf1 tale: function and regulation of an ubiquitin ligase in multiple cellular networks. Cell Mol Life Sci 2013; 70:2305-17. [PMID: 23007848 PMCID: PMC11113965 DOI: 10.1007/s00018-012-1170-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 09/05/2012] [Accepted: 09/11/2012] [Indexed: 01/17/2023]
Abstract
Since being discovered and intensively studied for over a decade, Smad ubiquitylation regulatory factor-1 (Smurf1) has been linked with several important biological pathways, including the bone morphogenetic protein pathway, the non-canonical Wnt pathway, and the mitogen-activated protein kinase pathway. Multiple functions of this ubiquitin ligase have been discovered in cell growth and morphogenesis, cell migration, cell polarity, and autophagy. Smurf1 is related to physiological manifestations in terms of age-dependent deficiency in bone formation and invasion of tumor cells. Smurf1-knockout mice have a significant phenotype in the skeletal system and considerable manifestations during embryonic development and neural outgrowth. In depth studying of Smurf1 will help us to understand the etiopathological mechanisms of related disorders. Here, we will summarize historical and recent studies on Smurf1, and discuss the E3 ligase-dependent and -independent functions of Smurf1. Moreover, intracellular regulations of Smurf1 and related physiological phenotypes will be described in this review.
Collapse
Affiliation(s)
- Yu Cao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850 China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850 China
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044 Liaoning Province China
| |
Collapse
|
32
|
Up-regulation of Smurf1 after spinal cord injury in adult rats. J Mol Histol 2013; 44:381-90. [PMID: 23595775 DOI: 10.1007/s10735-013-9499-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/12/2013] [Indexed: 01/16/2023]
|
33
|
Hwang YS, Lee HS, Kamata T, Mood K, Cho HJ, Winterbottom E, Ji YJ, Singh A, Daar IO. The Smurf ubiquitin ligases regulate tissue separation via antagonistic interactions with ephrinB1. Genes Dev 2013; 27:491-503. [PMID: 23475958 DOI: 10.1101/gad.208355.112] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The formation of tissue boundaries is dependent on the cell-cell adhesion/repulsion system that is required for normal morphogenetic processes during development. The Smad ubiquitin regulatory factors (Smurfs) are E3 ubiquitin ligases with established roles in cell growth and differentiation, but whose roles in regulating cell adhesion and migration are just beginning to emerge. Here, we demonstrate that the Smurfs regulate tissue separation at mesoderm/ectoderm boundaries through antagonistic interactions with ephrinB1, an Eph receptor ligand that has a key role in regulating the separation of embryonic germ layers. EphrinB1 is targeted by Smurf2 for degradation; however, a Smurf1 interaction with ephrinB1 prevents the association with Smurf2 and precludes ephrinB1 from ubiquitination and degradation, since it is a substantially weaker substrate for Smurf1. Inhibition of Smurf1 expression in embryonic mesoderm results in loss of ephrinB1-mediated separation of this tissue from the ectoderm, which can be rescued by the coincident inhibition of Smurf2 expression. This system of differential interactions between Smurfs and ephrinB1 regulates the maintenance of tissue boundaries through the control of ephrinB protein levels.
Collapse
Affiliation(s)
- Yoo-Seok Hwang
- Laboratory of Cell and Developmental Signaling, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The transforming growth factor β (TGFβ) superfamily of signal transduction molecules plays crucial roles in the regulation of cell behavior. TGFβ regulates gene transcription through Smad proteins and signals via non-Smad pathways. The TGFβ pathway is strictly regulated, and perturbations lead to tumorigenesis. Several pathway components are known to be targeted for proteasomal degradation via ubiquitination by E3 ligases. Smurfs are well known negative regulators of TGFβ, which function as E3 ligases recruited by adaptors such as I-Smads. TGFβ signaling can also be enhanced by E3 ligases, such as Arkadia, that target repressors for degradation. It is becoming clear that E3 ligases often target multiple pathways, thereby acting as mediators of signaling cross-talk. Regulation via ubiquitination involves a complex network of E3 ligases, adaptor proteins, and deubiquitinating enzymes (DUBs), the last-mentioned acting by removing ubiquitin from its targets. Interestingly, also non-degradative ubiquitin modifications are known to play important roles in TGFβ signaling. Ubiquitin modifications thus play a key role in TGFβ signal transduction, and in this review we provide an overview of known players, focusing on recent advances.
Collapse
Affiliation(s)
- Miriam De Boeck
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | | |
Collapse
|
35
|
Nethe M, de Kreuk BJ, Tauriello DVF, Anthony EC, Snoek B, Stumpel T, Salinas PC, Maurice MM, Geerts D, Deelder AM, Hensbergen PJ, Hordijk PL. Rac1 acts in conjunction with Nedd4 and dishevelled-1 to promote maturation of cell-cell contacts. J Cell Sci 2012; 125:3430-42. [PMID: 22467858 DOI: 10.1242/jcs.100925] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The Rho-GTPase Rac1 promotes actin polymerization and membrane protrusion that mediate initial contact and subsequent maturation of cell-cell junctions. Here we report that Rac1 associates with the ubiquitin-protein ligase neural precursor cell expressed developmentally down-regulated 4 (Nedd4). This interaction requires the hypervariable C-terminal domain of Rac1 and the WW domains of Nedd4. Activated Rac1 colocalises with endogenous Nedd4 at epithelial cell-cell contacts. Reduction of Nedd4 expression by shRNA results in reduced transepithelial electrical resistance (TER) and concomitant changes in the distribution of adherens and tight junction markers. Conversely, expression of Nedd4 promotes TER, suggesting that Nedd4 cooperates with Rac1 in the induction of junctional maturation. We found that Nedd4, but not Nedd4-2, mediates the ubiquitylation and degradation of the adapter protein dishevelled-1 (Dvl1), the expression of which negatively regulates cell-cell contact. Nedd4-mediated ubiquitylation requires its binding to the C-terminal domain of Dvl1, comprising the DEP domain, and targets an N-terminal lysine-rich region upstream of the Dvl1 DIX domain. We found that endogenous Rac1 colocalises with endogenous Dvl1 in intracellular puncta as well as on cell-cell junctions. Finally, activated Rac1 was found to stimulate Nedd4 activity, resulting in increased ubiquitylation of Dvl1. Together, these data reveal a novel Rac1-dependent signalling pathway that, through Nedd4-mediated ubiquitylation of Dvl1, stimulates the maturation of epithelial cell-cell contacts.
Collapse
Affiliation(s)
- Micha Nethe
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Image-based genome-wide siRNA screen identifies selective autophagy factors. Nature 2012; 480:113-7. [PMID: 22020285 PMCID: PMC3229641 DOI: 10.1038/nature10546] [Citation(s) in RCA: 400] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 12/01/2011] [Accepted: 09/07/2011] [Indexed: 12/25/2022]
Abstract
Selective autophagy involves the recognition and targeting of specific cargo, such as damaged organelles, misfolded proteins, or invading pathogens for lysosomal destruction. Yeast genetic screens have identified proteins required for different forms of selective autophagy, including cytoplasm-to-vacuole targeting, pexophagy and mitophagy, and mammalian genetic screens have identified proteins required for autophagy regulation. However, there have been no systematic approaches to identify molecular determinants of selective autophagy in mammalian cells. Here, to identify mammalian genes required for selective autophagy, we performed a high-content, image-based, genome-wide small interfering RNA screen to detect genes required for the colocalization of Sindbis virus capsid protein with autophagolysosomes. We identified 141 candidate genes required for viral autophagy, which were enriched for cellular pathways related to messenger RNA processing, interferon signalling, vesicle trafficking, cytoskeletal motor function and metabolism. Ninety-six of these genes were also required for Parkin-mediated mitophagy, indicating that common molecular determinants may be involved in autophagic targeting of viral nucleocapsids and autophagic targeting of damaged mitochondria. Murine embryonic fibroblasts lacking one of these gene products, the C2-domain containing protein, SMURF1, are deficient in the autophagosomal targeting of Sindbis and herpes simplex viruses and in the clearance of damaged mitochondria. Moreover, SMURF1-deficient mice accumulate damaged mitochondria in the heart, brain and liver. Thus, our study identifies candidate determinants of selective autophagy, and defines SMURF1 as a newly recognized mediator of both viral autophagy and mitophagy.
Collapse
|
37
|
Cell cycle arrest and apoptosis by expression of a novel TPIP (TPIP-C2) cDNA encoding a C2-domain in HEK-293 cells. Mol Biol Rep 2012; 39:7389-402. [PMID: 22311048 DOI: 10.1007/s11033-012-1571-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 01/25/2012] [Indexed: 12/15/2022]
Abstract
The human TPIP (TPTE and PTEN homologous Inositol lipid Phosphatase) belongs to the PTEN (Phosphatase and TENsin homologue deleted on chromosome 10) family of dual-specific phosphatases and is expressed from the human chromosome 13 as multiple splice-variants, e.g., TPIPα, β, γ mRNAs. PTEN is a well characterized tumor suppressor, which controls survival, adhesion, motility and migration of mammalian cells, its C2-domain plays crucial role in controlling these functions. However, role of isolated C2-domain protein in regulation of cell proliferation and apoptosis is not reported. We report sequence analysis and function of a novel human TPIP (TPIP-C2) cDNA encoding a 193 amino acid C2-domain in cell proliferation and apoptosis regulation. In silico analysis and homology modelling revealed that the C2-domain of TPIP-C2 is similar to that of PTEN but with short disorder sequences overlapping or adjacent to the post-translational modification sites. Overexpression of TPIP-C2 cDNA in human embryonic kidney (HEK-293) cells caused cell cycle arrest, inhibition of cell proliferation and induced apoptosis in an activated caspase 3 and PARP-dependent manner in comparison to overexpression of the full length human PTEN cDNA. TPIP-C2 overexpressed cells also showed S-phase cell cycle arrest. We suggest that C2-domain of TPIP-C2 may act as a dominant negative effector, which may bind to and arrest the cell proliferation signalling complex and isolated TPIP-C2-domain-like proteins expressed in mammalian cells/tissues may play important role in regulation of cell proliferation and apoptosis. The TPIP-C2 cDNA may be exploited for inducing cell cycle-inhibition and apoptosis in human cancer cells and tissues.
Collapse
|
38
|
Soond SM, Chantry A. How ubiquitination regulates the TGF-β signalling pathway: new insights and new players: new isoforms of ubiquitin-activating enzymes in the E1-E3 families join the game. Bioessays 2012; 33:749-58. [PMID: 21932223 DOI: 10.1002/bies.201100057] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ubiquitination of protein species in regulating signal transduction pathways is universally accepted as of fundamental importance for normal development, and defects in this process have been implicated in the progression of many human diseases. One pathway that has received much attention in this context is transforming growth factor-beta (TGF-β) signalling, particularly during the regulation of epithelial-mesenchymal transition (EMT) and tumour progression. While E3-ubiquitin ligases offer themselves as potential therapeutic targets, much remains to be unveiled regarding mechanisms that culminate in their regulation. With this in mind, the focus of this review highlights the regulation of the ubiquitination pathway and the significance of a recently described group of NEDD4 E3-ubiquitin ligase isoforms in the context of TGF-β pathway regulation. Moreover, we now broaden these observations to incorporate a growing number of protein isoforms within the ubiquitin ligase superfamily as a whole, and discuss their relevance in defining a new 'iso-ubiquitinome'.
Collapse
Affiliation(s)
- Surinder M Soond
- University of East Anglia, School Of Biological Sciences, Norwich, Norfolk, UK.
| | | |
Collapse
|
39
|
Farooqi AA, Waseem MS, Riaz AM, Bhatti S. SMURF and NEDD4: sharp shooters monitor the gate keepers and ion traffic controllers of lead astray cell. J Membr Biol 2011; 244:1-8. [PMID: 21918841 DOI: 10.1007/s00232-011-9394-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 08/29/2011] [Indexed: 10/17/2022]
Abstract
It is becoming increasingly apparent that a complex bar code underlies the quantitative aspects of extracellular signal regulation. Cell type-specific and context-dependent transcriptional programs are triggered by sophisticated nanomachinery consisting of HECT enzymes which monitor signal generation, transduction and termination. How the HECT enzymes safeguard spatiotemporal organization was a fundamental question towards understanding the process of protein degradation and its functions in diverse biological processes. In this review we will dismantle how HECT E3 enzymes regulate the trafficking of many receptors, channels and transporters as well as how HECT enzymes negatively regulate each other. There is accumulating evidence that suggests an undeniable role of HECT enzymes in regulating mediators of the Wnt signal-transduction cascade. By contrast, little is known about the crosstalk of HECT enzymes with ATM and TRAIL in prostate cancer, but several hints have emerged. This review provides a broader snapshot for studying multiple pathways in parallel, rather than as separate entities.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan.
| | | | | | | |
Collapse
|