1
|
Ho HH, Wing SS. α-Synuclein ubiquitination - functions in proteostasis and development of Lewy bodies. Front Mol Neurosci 2024; 17:1498459. [PMID: 39600913 PMCID: PMC11588729 DOI: 10.3389/fnmol.2024.1498459] [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: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Synucleinopathies are neurodegenerative disorders characterized by the accumulation of α-synuclein containing Lewy bodies. Ubiquitination, a key post-translational modification, has been recognized as a pivotal regulator of α-synuclein's cellular dynamics, influencing its degradation, aggregation, and associated neurotoxicity. This review examines comprehensively the current understanding of α-synuclein ubiquitination and its role in the pathogenesis of synucleinopathies, particularly in the context of Parkinson's disease. We explore the molecular mechanisms responsible for α-synuclein ubiquitination, with a focus on the roles of E3 ligases and deubiquitinases implicated in the degradation process which occurs primarily through the endosomal lysosomal pathway. The review further discusses how the dysregulation of these mechanisms contributes to α-synuclein aggregation and LB formation and offers suggestions for future investigations into the role of α-synuclein ubiquitination. Understanding these processes may shed light on potential therapeutic avenues that can modulate α-synuclein ubiquitination to alleviate its pathological impact in synucleinopathies.
Collapse
Affiliation(s)
- Hung-Hsiang Ho
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Simon S. Wing
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Jaykumar AB, Plumber S, Binns D, Wichaidit C, Luby-Phelps K, Cobb MH. SMURF1/2 are novel regulators of WNK1 stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606092. [PMID: 39131382 PMCID: PMC11312594 DOI: 10.1101/2024.07.31.606092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Angiogenesis is essential for remodeling and repairing existing vessels, and this process requires signaling pathways including those controlled by transforming growth factor beta (TGF-β). We have previously reported crosstalk between TGF-β and the protein kinase With No lysine (K) 1 (WNK1). Homozygous disruption of the gene encoding WNK1 results in lethality in mice near embryonic day E12 due to impaired angiogenesis and this defect can be rescued by endothelial-specific expression of an activated form of the WNK1 substrate kinase Oxidative Stress-Responsive 1 (OSR1). However, molecular processes regulated via a collaboration between TGF-β and WNK1/OSR1 are not well understood. Here we show that WNK1 interacts with the E3 ubiquitin ligases SMURF1/2. In addition, we discovered that WNK1 regulates SMURF1/2 protein stability and vice versa. We also demonstrate that WNK1 activity regulates TGF-β receptor levels, in turn, controlling TGF-β signaling.
Collapse
Affiliation(s)
| | - Sakina Plumber
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Derk Binns
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | | | | | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| |
Collapse
|
3
|
Chen R, Kang Z, Li W, Xu T, Wang Y, Jiang Q, Wang Y, Huang Z, Xu X, Huang Z. Extracellular vesicle surface display of αPD-L1 and αCD3 antibodies via engineered late domain-based scaffold to activate T-cell anti-tumor immunity. J Extracell Vesicles 2024; 13:e12490. [PMID: 39051742 PMCID: PMC11270581 DOI: 10.1002/jev2.12490] [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: 12/25/2023] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Extracellular vesicles (EVs) are emerging as promising carriers for the delivery of therapeutic biologics. Genetic engineering represents a robust strategy for loading proteins of interest into EVs. Identification of EV-enriched proteins facilitates protein cargo loading efficiency. Many EV-enriched proteins are sorted into EVs via an endosomal sorting complex required for transport (ESCRT)-dependent pathway. In parallel, viruses hijack this EV biosynthesis machinery via conserved late domain motifs to promote egress from host cells. Inspired by the similarity of biogenesis between EVs and viruses, we developed a synthetic, Late domain-based EV scaffold protein that enables the display of a set of single chain variable fragments (scFvs) on the EV surface. We named this scaffold the Late domain-based exosomal antibody surface display platform (LEAP). We applied the LEAP scaffold to reprogramme HEK293T cell-derived EVs to elicit T-cell anti-tumor immunity by simultaneously displaying αPD-L1 and αCD3 scFvs on the EV surface (denoted as αPD-L1×αCD3 bispecific T-cell engaging exosomes, BiTExos). We demonstrated that αPD-L1×αCD3 BiTExos actively redirected T cells to bind to PD-L1+ tumor cells, promoting T-cell activation, proliferation and tumoricidal cytokine production. Furthermore, the αPD-L1×αCD3 BiTExos promoted T-cell infiltration into the tumor microenvironment to mitigate the tumor burden in vivo. Our study suggested that the LEAP scaffold may serve as a platform for EV surface display and could be applied for a broad range of EV-based biomedical applications.
Collapse
Affiliation(s)
- Rui Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Ziqin Kang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Wenhao Li
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Tianshu Xu
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Yongqiang Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Medical Research CenterSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Qiming Jiang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Yuepeng Wang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Zixian Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongP.R. China
- Medical Research CenterSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| | - Zhiquan Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP.R. China
| |
Collapse
|
4
|
Wedegaertner H, Bosompra O, Kufareva I, Trejo J. Divergent regulation of α-arrestin ARRDC3 function by ubiquitination. Mol Biol Cell 2023; 34:ar93. [PMID: 37223976 PMCID: PMC10398895 DOI: 10.1091/mbc.e23-02-0055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/14/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023] Open
Abstract
The α-arrestin ARRDC3 is a recently discovered tumor suppressor in invasive breast cancer that functions as a multifaceted adaptor protein to control protein trafficking and cellular signaling. However, the molecular mechanisms that control ARRDC3 function are unknown. Other arrestins are known to be regulated by posttranslational modifications, suggesting that ARRDC3 may be subject to similar regulatory mechanisms. Here we report that ubiquitination is a key regulator of ARRDC3 function and is mediated primarily by two proline-rich PPXY motifs in the ARRDC3 C-tail domain. Ubiquitination and the PPXY motifs are essential for ARRDC3 function in regulating GPCR trafficking and signaling. Additionally, ubiquitination and the PPXY motifs mediate ARRDC3 protein degradation, dictate ARRDC3 subcellular localization, and are required for interaction with the NEDD4-family E3 ubiquitin ligase WWP2. These studies demonstrate a role for ubiquitination in regulating ARRDC3 function and reveal a mechanism by which ARRDC3 divergent functions are controlled.
Collapse
Affiliation(s)
- Helen Wedegaertner
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA92093
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA92093
| | - Oye Bosompra
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA92093
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA92093
| | - Irina Kufareva
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA92093
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA92093
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA92093
| |
Collapse
|
5
|
Kennedy A, Waters E, Rowshanravan B, Hinze C, Williams C, Janman D, Fox TA, Booth C, Pesenacker AM, Halliday N, Soskic B, Kaur S, Qureshi OS, Morris EC, Ikemizu S, Paluch C, Huo J, Davis SJ, Boucrot E, Walker LSK, Sansom DM. Differences in CD80 and CD86 transendocytosis reveal CD86 as a key target for CTLA-4 immune regulation. Nat Immunol 2022; 23:1365-1378. [PMID: 35999394 PMCID: PMC9477731 DOI: 10.1038/s41590-022-01289-w] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/15/2022] [Indexed: 01/07/2023]
Abstract
CD28 and CTLA-4 (CD152) play essential roles in regulating T cell immunity, balancing the activation and inhibition of T cell responses, respectively. Although both receptors share the same ligands, CD80 and CD86, the specific requirement for two distinct ligands remains obscure. In the present study, we demonstrate that, although CTLA-4 targets both CD80 and CD86 for destruction via transendocytosis, this process results in separate fates for CTLA-4 itself. In the presence of CD80, CTLA-4 remained ligand bound, and was ubiquitylated and trafficked via late endosomes and lysosomes. In contrast, in the presence of CD86, CTLA-4 detached in a pH-dependent manner and recycled back to the cell surface to permit further transendocytosis. Furthermore, we identified clinically relevant mutations that cause autoimmune disease, which selectively disrupted CD86 transendocytosis, by affecting either CTLA-4 recycling or CD86 binding. These observations provide a rationale for two distinct ligands and show that defects in CTLA-4-mediated transendocytosis of CD86 are associated with autoimmunity.
Collapse
Affiliation(s)
- Alan Kennedy
- UCL Institute of Immunity and Transplantation, London, UK
| | - Erin Waters
- UCL Institute of Immunity and Transplantation, London, UK
| | | | - Claudia Hinze
- UCL Institute of Immunity and Transplantation, London, UK
| | | | - Daniel Janman
- UCL Institute of Immunity and Transplantation, London, UK
| | - Thomas A Fox
- UCL Institute of Immunity and Transplantation, London, UK
| | - Claire Booth
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Neil Halliday
- UCL Institute of Immunity and Transplantation, London, UK
| | - Blagoje Soskic
- UCL Institute of Immunity and Transplantation, London, UK
| | - Satdip Kaur
- School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, UK
| | | | - Emma C Morris
- UCL Institute of Immunity and Transplantation, London, UK
| | - Shinji Ikemizu
- Division of Structural Biology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Christopher Paluch
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jiandong Huo
- Structural Biology, The Rosalind Franklin Institute, Didcot, UK
- Division of Structural Biology, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, Oxford, UK
- Protein Production UK, The Rosalind Franklin Institute-Diamond Light Source, The Research Complex at Harwell, Didcot, UK
| | - Simon J Davis
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, University College London, London, UK
| | | | - David M Sansom
- UCL Institute of Immunity and Transplantation, London, UK.
| |
Collapse
|
6
|
Zhang Q, Gong W, Wu H, Wang J, Jin Q, Lin C, Xu S, Bao W, Wang Y, Wu J, Feng S, Zhao C, Chen B, Liu Z. DKK1 suppresses WWP2 to enhance bortezomib resistance in multiple myeloma via regulating GLI2 ubiquitination. Carcinogenesis 2021; 42:1223-1231. [PMID: 34546340 DOI: 10.1093/carcin/bgab086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/13/2021] [Accepted: 09/17/2021] [Indexed: 11/14/2022] Open
Abstract
Bortezomib-based chemotherapy represents the most prevalent regimens for multiple myeloma (MM), whereas acquired drug resistance remains a major obstacle. Myeloma cells often produce excessive amount of dickkopf-1 (DKK1), giving rise to myeloma bone disease. However, it remains obscure about the effects and mechanisms of DKK1 in the progression and bortezomib responsiveness of MM cells. In the current study, we found WWP2, an E3 ubiquitin-protein ligase, was downregulated in the bortezomib-resistant cells along with high expression of DKK1. Further investigation revealed that WWP2 was a direct target of Wnt/β-catenin signaling pathway, and DKK1 suppressed the expression of WWP2 via canonical Wnt signaling. We further identified that WWP2 mediated the ubiquitination and degradation of GLI2, a main transcriptional factor of the Hedgehog (Hh) pathway. Therefore, DKK1-induced WWP2 downregulation improved GLI2 stability and activation of Hh signaling pathway, contributing to the resistance to bortezomib of MM cells. Clinical data also validated that WWP2 expression was associated with the treatment response and clinic outcomes of MM patients. WWP2 overexpression restricted MM progression and enhanced cell sensitivity to bortezomib treatment in vitro and in vivo. Taken together, our findings demonstrate that DKK1 facilitates the generation of bortezomib resistance in MM via downregulating WWP2 and activating Hh pathway. Thus, the manipulation of DKK1-WWP2-GLI2 axis might sensitize myeloma cells to proteasome inhibitors.
Collapse
Affiliation(s)
- Qiguo Zhang
- Department of Hematology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu, China.,Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Wenyu Gong
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Hongyan Wu
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu, China
| | - Jing Wang
- Department of Hematology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu, China
| | - Qichuan Jin
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Chun Lin
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Shiyun Xu
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Wenqiang Bao
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Yin Wang
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Jing Wu
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Shanshan Feng
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Changzhi Zhao
- Department of Hematology, Chuzhou First People's Hospital of Anhui Medical University, Chuzhou, Anhui
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu, China
| | - Zhiqiang Liu
- Department of Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| |
Collapse
|
7
|
Predicting PY motif-mediated protein-protein interactions in the Nedd4 family of ubiquitin ligases. PLoS One 2021; 16:e0258315. [PMID: 34637467 PMCID: PMC8509885 DOI: 10.1371/journal.pone.0258315] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023] Open
Abstract
The Nedd4 family contains several structurally related but functionally distinct HECT-type ubiquitin ligases. The members of the Nedd4 family are known to recognize substrates through their multiple WW domains, which recognize PY motifs (PPxY, LPxY) or phospho-threonine or phospho-serine residues. To better understand protein interactor recognition mechanisms across the Nedd4 family, we report the development and implementation of a python-based tool, PxYFinder, to identify PY motifs in the primary sequences of previously identified interactors of Nedd4 and related ligases. Using PxYFinder, we find that, on average, half of Nedd4 family interactions are likely PY-motif mediated. Further, we find that PPxY motifs are more prevalent than LPxY motifs and are more likely to occur in proline-rich regions and that PPxY regions are more disordered on average relative to LPxY-containing regions. Informed by consensus sequences for PY motifs across the Nedd4 interactome, we rationally designed a focused peptide library and employed a computational screen, revealing sequence- and biomolecular interaction-dependent determinants of WW-domain/PY-motif interactions. Cumulatively, our efforts provide a new bioinformatic tool and expand our understanding of sequence and structural factors that contribute to PY-motif mediated interactor recognition across the Nedd4 family.
Collapse
|
8
|
TcpC inhibits neutrophil extracellular trap formation by enhancing ubiquitination mediated degradation of peptidylarginine deiminase 4. Nat Commun 2021; 12:3481. [PMID: 34108482 PMCID: PMC8190435 DOI: 10.1038/s41467-021-23881-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 05/24/2021] [Indexed: 01/21/2023] Open
Abstract
TcpC is a multifunctional virulence factor of uropathogenic E. coli (UPEC). Neutrophil extracellular trap formation (NETosis) is a crucial anti-infection mechanism of neutrophils. Here we show the influence of TcpC on NETosis and related mechanisms. We show NETosis in the context of a pyelonephritis mouse model induced by TcpC-secreting wild-type E. coli CFT073 (CFT073wt) and LPS-induced in vitro NETosis with CFT073wt or recombinant TcpC (rTcpC)-treated neutrophils are inhibited. rTcpC enters neutrophils through caveolin-mediated endocytosis and inhibits LPS-induced production of ROS, proinflammatory cytokines and protein but not mRNA levels of peptidylarginine deiminase 4 (PAD4). rTcpC treatment enhances PAD4 ubiquitination and accumulation in proteasomes. Moreover, in vitro ubiquitination kit analyses show that TcpC is a PAD4-targetd E3 ubiquitin-ligase. These data suggest that TcpC inhibits NETosis primarily by serving as an E3 ligase that promotes degradation of PAD4. Our findings provide a novel mechanism underlying TcpC-mediated innate immune evasion.
Collapse
|
9
|
TcpC inhibits toll-like receptor signaling pathway by serving as an E3 ubiquitin ligase that promotes degradation of myeloid differentiation factor 88. PLoS Pathog 2021; 17:e1009481. [PMID: 33788895 PMCID: PMC8041205 DOI: 10.1371/journal.ppat.1009481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/12/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
TcpC is a virulence factor of uropathogenic E. coli (UPEC). It was found that TIR domain of TcpC impedes TLR signaling by direct association with MyD88. It has been a long-standing question whether bacterial pathogens have evolved a mechanism to manipulate MyD88 degradation by ubiquitin-proteasome pathway. Here, we show that TcpC is a MyD88-targeted E3 ubiquitin ligase. Kidney macrophages from mice with pyelonephritis induced by TcpC-secreting UPEC showed significantly decreased MyD88 protein levels. Recombinant TcpC (rTcpC) dose-dependently inhibited protein but not mRNA levels of MyD88 in macrophages. Moreover, rTcpC significantly promoted MyD88 ubiquitination and accumulation in proteasomes in macrophages. Cys12 and Trp106 in TcpC are crucial amino acids in maintaining its E3 activity. Therefore, TcpC blocks TLR signaling pathway by degradation of MyD88 through ubiquitin-proteasome system. Our findings provide not only a novel biochemical mechanism underlying TcpC-medicated immune evasion, but also the first example that bacterial pathogens inhibit MyD88-mediated signaling pathway by virulence factors that function as E3 ubiquitin ligase. Toll/interleukin-1 receptor domain-containing protein encoded by E. coli (TcpC) is an important virulence factor in many strains of uropathogenic E. coli (UPEC). TcpC-mediated evasion of innate immunity plays an important role in the pathogenesis of UPEC caused urinary tract infection (UTI) including pyelonephritis. In the present study, we show TcpC is an E3 ubiquitin ligase that promotes ubiquitination and degradation of MyD88, hereby blocking the TLR signaling pathway. Our findings not only illuminate the novel biochemical mechanisms underlying TcpC-mediated evasion of innate immunity, but also provide the first example that bacterial pathogens can subvert TLR signaling pathway through virulence factors that function as MyD88-targeted E3 ubiquitin ligase.
Collapse
|
10
|
Zeng J, Santos AF, Mukadam AS, Osswald M, Jacques DA, Dickson CF, McLaughlin SH, Johnson CM, Kiss L, Luptak J, Renner N, Vaysburd M, McEwan WA, Morais-de-Sá E, Clift D, James LC. Target-induced clustering activates Trim-Away of pathogens and proteins. Nat Struct Mol Biol 2021; 28:278-289. [PMID: 33633400 PMCID: PMC7611929 DOI: 10.1038/s41594-021-00560-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023]
Abstract
Trim-Away is a recently developed technology that exploits off-the-shelf antibodies and the RING E3 ligase and cytosolic antibody receptor TRIM21 to carry out rapid protein depletion. How TRIM21 is catalytically activated upon target engagement, either during its normal immune function or when repurposed for targeted protein degradation, is unknown. Here we show that a mechanism of target-induced clustering triggers intermolecular dimerization of the RING domain to switch on the ubiquitination activity of TRIM21 and induce virus neutralization or drive Trim-Away. We harness this mechanism for selective degradation of disease-causing huntingtin protein containing long polyglutamine tracts and expand the Trim-Away toolbox with highly active TRIM21-nanobody chimeras that can also be controlled optogenetically. This work provides a mechanism for cellular activation of TRIM RING ligases and has implications for targeted protein degradation technologies.
Collapse
Affiliation(s)
- Jingwei Zeng
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Ana Filipa Santos
- i3S - Instituto de Investigação e Inovação em Saúde and IBMC Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Aamir S. Mukadam
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mariana Osswald
- i3S - Instituto de Investigação e Inovação em Saúde and IBMC Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - David A. Jacques
- EMBL Australia Node, Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Claire F. Dickson
- EMBL Australia Node, Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | | | - Leo Kiss
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Jakub Luptak
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Nadine Renner
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Marina Vaysburd
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - William A. McEwan
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| | - Eurico Morais-de-Sá
- i3S - Instituto de Investigação e Inovação em Saúde and IBMC Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| | - Dean Clift
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| | - Leo C. James
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| |
Collapse
|
11
|
Adaptors as the regulators of HECT ubiquitin ligases. Cell Death Differ 2021; 28:455-472. [PMID: 33402750 DOI: 10.1038/s41418-020-00707-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/04/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022] Open
Abstract
The HECT (homologous to E6AP C-terminus) ubiquitin ligases (E3s) are a small family of highly conserved enzymes involved in diverse cellular functions and pathological conditions. Characterised by a C-terminal HECT domain that accepts ubiquitin from E2 ubiquitin conjugating enzymes, these E3s regulate key signalling pathways. The activity and functional regulation of HECT E3s are controlled by several factors including post-translational modifications, inter- and intramolecular interactions and binding of co-activators and adaptor proteins. In this review, we focus on the regulation of HECT E3s by accessory proteins or adaptors and discuss various ways by which adaptors mediate their regulatory roles to affect physiological outcomes. We discuss common features that are conserved from yeast to mammals, regardless of the type of E3s as well as shed light on recent discoveries explaining some existing enigmas in the field.
Collapse
|
12
|
Dimou S, Diallinas G. Life and Death of Fungal Transporters under the Challenge of Polarity. Int J Mol Sci 2020; 21:ijms21155376. [PMID: 32751072 PMCID: PMC7432044 DOI: 10.3390/ijms21155376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
Eukaryotic plasma membrane (PM) transporters face critical challenges that are not widely present in prokaryotes. The two most important issues are proper subcellular traffic and targeting to the PM, and regulated endocytosis in response to physiological, developmental, or stress signals. Sorting of transporters from their site of synthesis, the endoplasmic reticulum (ER), to the PM has been long thought, but not formally shown, to occur via the conventional Golgi-dependent vesicular secretory pathway. Endocytosis of specific eukaryotic transporters has been studied more systematically and shown to involve ubiquitination, internalization, and sorting to early endosomes, followed by turnover in the multivesicular bodies (MVB)/lysosomes/vacuole system. In specific cases, internalized transporters have been shown to recycle back to the PM. However, the mechanisms of transporter forward trafficking and turnover have been overturned recently through systematic work in the model fungus Aspergillus nidulans. In this review, we present evidence that shows that transporter traffic to the PM takes place through Golgi bypass and transporter endocytosis operates via a mechanism that is distinct from that of recycling membrane cargoes essential for fungal growth. We discuss these findings in relation to adaptation to challenges imposed by cell polarity in fungi as well as in other eukaryotes and provide a rationale of why transporters and possibly other housekeeping membrane proteins ‘avoid’ routes of polar trafficking.
Collapse
|
13
|
Diallinas G, Martzoukou O. Transporter membrane traffic and function: lessons from a mould. FEBS J 2019; 286:4861-4875. [DOI: 10.1111/febs.15078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/26/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Affiliation(s)
- George Diallinas
- Department of Biology National and Kapodistrian University of Athens Greece
| | - Olga Martzoukou
- Department of Biology National and Kapodistrian University of Athens Greece
| |
Collapse
|
14
|
Abstract
Cell nutrition, detoxification, signalling, homeostasis and response to drugs, processes related to cell growth, differentiation and survival are all mediated by plasma membrane (PM) proteins called transporters. Despite their distinct fine structures, mechanism of function, energetic requirements, kinetics and substrate specificities, all transporters are characterized by a main hydrophobic body embedded in the PM as a series of tightly packed, often intertwined, α-helices that traverse the lipid bilayer in a zigzag mode, connected with intracellular or extracellular loops and hydrophilic N- and C-termini. Whereas longstanding genetic, biochemical and biophysical evidence suggests that specific transmembrane segments, and also their connecting loops, are responsible for substrate recognition and transport dynamics, emerging evidence also reveals the functional importance of transporter N- and C-termini, in respect to transport catalysis, substrate specificity, subcellular expression, stability and signalling. This review highlights selected prototypic examples of transporters in which their termini play important roles in their functioning.
Collapse
Affiliation(s)
- Emmanuel Mikros
- Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, 15771 Athens, Greece
| | - George Diallinas
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15781 Athens, Greece
| |
Collapse
|
15
|
Yao W, Shan Z, Gu A, Fu M, Shi Z, Wen W. WW domain-mediated regulation and activation of E3 ubiquitin ligase Suppressor of Deltex. J Biol Chem 2018; 293:16697-16708. [PMID: 30213861 DOI: 10.1074/jbc.ra118.003781] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/27/2018] [Indexed: 12/22/2022] Open
Abstract
The Nedd4 family E3 ligases Itch and WWP1/2 play crucial roles in the regulation of cell cycle progression and apoptosis and are closely correlated with cancer development and metastasis. It has been recently shown that the ligase activities of Itch and WWP1/2 are tightly regulated, with the HECT domain sequestered intramolecularly by a linker region connecting WW2 and WW3. Here, we show that a similar autoinhibitory mechanism is utilized by the Drosophila ortholog of Itch and WWP1/2, Suppressor of Deltex (Su(dx)). We show that Su(dx) adopts an inactive steady state with the WW domain region interacting with the HECT domain. We demonstrate that both the linker and preceding WW2 are required for the efficient binding and regulation of Su(dx) HECT. Recruiting the multiple-PY motif-containing adaptor dNdfip via WW domains relieves the inhibitory state of Su(dx) and leads to substrate (e.g. Notch) ubiquitination. Our study demonstrates an evolutionarily conservative mechanism governing the regulation and activation of some Nedd4 family E3 ligases. Our results also suggest a dual regulatory mechanism for specific Notch down-regulation via dNdfip-Su(dx)-mediated Notch ubiquitination.
Collapse
Affiliation(s)
- Weiyi Yao
- From the Department of Neurosurgery, Huashan Hospital, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China and
| | - Zelin Shan
- From the Department of Neurosurgery, Huashan Hospital, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China and
| | - Aihong Gu
- From the Department of Neurosurgery, Huashan Hospital, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China and
| | - Minjie Fu
- From the Department of Neurosurgery, Huashan Hospital, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China and
| | - Zhifeng Shi
- From the Department of Neurosurgery, Huashan Hospital, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China and
| | - Wenyu Wen
- From the Department of Neurosurgery, Huashan Hospital, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200040, China and .,the Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Mund T, Masuda-Suzukake M, Goedert M, Pelham HR. Ubiquitination of alpha-synuclein filaments by Nedd4 ligases. PLoS One 2018; 13:e0200763. [PMID: 30021006 PMCID: PMC6051637 DOI: 10.1371/journal.pone.0200763] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/02/2018] [Indexed: 11/19/2022] Open
Abstract
Alpha-synuclein can form beta-sheet filaments, the accumulation of which plays a key role in the development of Parkinson’s disease, dementia with Lewy bodies and multiple system atrophy. It has previously been shown that alpha-synuclein is a substrate for the HECT domain-containing ubiquitin ligase Nedd4, and is subject to ubiquitin-mediated endosomal degradation. We show here that alpha-synuclein filaments are much better substrates for ubiquitination in vitro than monomeric alpha-synuclein, and that this increased susceptibility cannot be mimicked by the mere clustering of monomers. Recognition by Nedd4 family enzymes is not through the conventional binding of PPxY-containing sequences to WW domains of the ligase, but it also involves C2 and HECT domains. The disease-causing alpha-synuclein mutant A53T is a much less efficient substrate for Nedd4 ligases than the wild-type protein. We suggest that preferential recognition, ubiquitination and degradation of beta-sheet-containing filaments may help to limit toxicity, and that A53T alpha-synuclein may be more toxic, at least in part because it avoids this fate.
Collapse
Affiliation(s)
- Thomas Mund
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | - Michel Goedert
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Hugh R. Pelham
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|