1
|
Park S, Takwale AD, Lee JE, Yu N, Kim YH, Lee JY, Cho YH, Cho JH, Moon JH, Lee G, Kim JA, Hwang JY, Kim JH. Discovery of a novel molecular glue degrader targeting GSPT1/2 with a non-IMiD-based CRBN binder. Eur J Med Chem 2025; 291:117642. [PMID: 40252382 DOI: 10.1016/j.ejmech.2025.117642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Targeting undruggable proteins by inducing proximity between E3 ligase and their substrates has emerged as an innovative strategy for tackling challenging diseases. In this study, we identified a novel GSPT1 degrader, 4a (KMG-1068), through screening of our in-house small molecule library. Treatment with 4a demonstrated significant anti-proliferative activity across multiple cell lines, which was diminished by co-treatment with MLN4924, suggesting the involvement of the Cullin-containing complex. Quantitative proteomic analysis indicated that 4a predominantly induces the degradation of GSPT1/2. We further validated that 4a-mediated GSPT1/2 degradation is dependent on both CUL4 and CRBN. Moreover, 4a forms a ternary complex with CRBN and GSPT1/2, albeit with weaker binding affinity compared to reported GSPT1 molecular glues. BRET assays and competition assays with pomalidomide demonstrated that 4a binds to the C-terminal IMiD binding site of CRBN, leading to the degradation of GSPT1. Despite lacking the characteristic glutarimide moiety present in other CRBN-based molecular glue degraders, 4a interacts effectively with the IMiD binding site of CRBN. Structural characterization through analog synthesis further underscored the importance of specific structural features for CRBN engagement, GSPT1/2 degradation, and anti-proliferative effects, establishing 4a as a promising novel GSPT1/2 degrader with significant therapeutic potential.
Collapse
Affiliation(s)
- Seulki Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Akshay D Takwale
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 305-606, Republic of Korea
| | - Ji-Eun Lee
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea; Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Namsik Yu
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 305-606, Republic of Korea
| | - Yong Hyo Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 305-606, Republic of Korea
| | - Joo Youn Lee
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 305-606, Republic of Korea
| | - Yong Hee Cho
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 305-606, Republic of Korea; Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jin Hwa Cho
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jeong Hee Moon
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Gaseul Lee
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jung-Ae Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea; Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jong Yeon Hwang
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 305-606, Republic of Korea; Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Jeong-Hoon Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
2
|
Hussain MS, Eltaib L, Rana AJ, Maqbool M, Ashique S, Alanazi MN, Khan Y, Agrawal M. Exploiting E3 ligases for lung cancer therapy: The promise of DCAF-PROTACs. Pathol Res Pract 2025; 270:156001. [PMID: 40359818 DOI: 10.1016/j.prp.2025.156001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
Lung cancer remains the leading cause of cancer-related mortality, underscoring the urgent need for novel therapeutic strategies. One emerging approach in drug development targets oncogenic proteins via the ubiquitin-proteasome system (UPS), specifically through proteolysis-targeting chimeras (PROTACs). Among the various E3 ligase complexes, the CRL4 complex-comprising DDB1 and CUL4-associated factors (DCAFs)-has garnered attention for its roles in cellular homeostasis, DNA repair, and oncogenesis. This review explores the therapeutic potential of DCAF-based PROTACs (DCAF-PROTACs) in lung cancer by focusing on the substrate receptors DCAF13, DCAF15, and DCAF16, which mediate CRL4-dependent ubiquitination. We first discuss the dysregulation of DCAF proteins in lung cancer and then elaborate on their mechanistic role in facilitating target-specific protein degradation via DCAF-E3 ligase complexes. Recent studies show that DCAF-PROTACs selectively degrade oncogenic proteins, addressing treatment resistance and tumor heterogeneity. Notably, DCAF13 promotes lung adenocarcinoma by destabilizing p53, while DCAF15-PROTACs target and degrade RBM39 effectively. Additionally, the development of electrophilic PROTACs targeting DCAF16 presents a promising avenue for degrading nuclear proteins. Despite these advancements, several challenges must be addressed prior to clinical translation, including issues related to drug bioavailability, stability, and emerging resistance mechanisms. This review also explores the potential of combination therapies, particularly with immunotherapy, to enhance tumor specificity and therapeutic efficacy. Ultimately, the deployment of DCAF-PROTACs marks a significant advancement in precision oncology, offering a novel and targeted approach to protein degradation-based cancer treatment.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand 248007, India.
| | - Lina Eltaib
- Department of Pharmaceutics, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Amita Joshi Rana
- College of Pharmacy, Graphic Era Hill University, Bhimtal, Uttarakhand 263136, India
| | - Mudasir Maqbool
- Department of Pharmacology, Government Medical College Baramulla, Jammu and Kashmir 193103, India
| | - Sumel Ashique
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, West Bengal 711316, India
| | - Mashael N Alanazi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Yumna Khan
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture, Peshawar, Khyber Pakhtunkhwa 25000, Pakistan
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram 122103, India
| |
Collapse
|
3
|
Wootton LM, Morgan EL. Ubiquitin and ubiquitin-like proteins in HPV-driven carcinogenesis. Oncogene 2025; 44:713-723. [PMID: 40011575 PMCID: PMC11888991 DOI: 10.1038/s41388-025-03310-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025]
Abstract
Persistent infection with high-risk (HR) human papillomaviruses (HPVs) is responsible for approximately 5% of cancer cases worldwide, including a growing number of oropharyngeal and anogenital cancers. The major HPV oncoproteins, E6 and E7, act together to manipulate cellular pathways involved in the regulation of proliferation, the cell cycle and cell survival, ultimately driving malignant transformation. Protein ubiquitination and the ubiquitin proteasome system (UPS) is often deregulated upon viral infection and in oncogenesis. HPV E6 and E7 interact with and disrupt multiple components of the ubiquitination machinery to promote viral persistence, which can also result in cellular transformation and the formation of tumours. This review highlights the ways in which HPV manipulates protein ubiquitination and the ubiquitin-like protein pathways and how this contributes to tumour development. Furthermore, we discuss how understanding the interactions between HPV and the protein ubiquitination could lead to novel therapeutic targets that are of urgent need in HPV+ carcinomas.
Collapse
Affiliation(s)
| | - Ethan L Morgan
- School of Life Sciences, University of Sussex, Brighton, UK.
| |
Collapse
|
4
|
Burgess AE, Loughran TA, Turk LS, Nyvall HG, Dunlop JL, Jamieson SA, Curry JR, Burke JE, Filipcik P, Brown SHJ, Mace PD. DET1 dynamics underlie cooperative ubiquitination by CRL4 DET1-COP1 complexes. SCIENCE ADVANCES 2025; 11:eadq4187. [PMID: 40009677 PMCID: PMC11864197 DOI: 10.1126/sciadv.adq4187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Transcription factor ubiquitination is a decisive regulator of growth and development. The DET1-DDB1-DDA1 (DDD) complex associates with the Cullin-4 ubiquitin ligase (CRL4) and a second ubiquitin ligase, COP1, to control ubiquitination of transcription factors involved in neurological, metabolic, and immune cell development. Here, we report the structure of the human DDD complex, revealing a specific segment of DET1 that can recruit ubiquitin-conjugating (E2) enzymes. Structural variability analysis, mass spectrometry, and mutagenesis based on AlphaFold predictions suggest that dynamic closure of DET1, stabilized by DDA1, underlies coordinated recruitment of E2 enzymes and COP1. Biochemical assays suggest that the E2 acts as a recruitment factor to bring COP1 to DET1 for more effective substrate ubiquitination, which parallels a catalytically inactive E2 enzyme (COP10) in plant DDD complexes. This work provides a clear architecture for regulation and cooperative CRL4DET1-COP1 complex assembly, which can affect degradation of diverse targets by COP1 complexes.
Collapse
Affiliation(s)
- Abigail E. Burgess
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Tarren A. Loughran
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Liam S. Turk
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Hunter G. Nyvall
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Jessica L. Dunlop
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sam A. Jamieson
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Jack R. Curry
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - John E. Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Pavel Filipcik
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Simon H. J. Brown
- School of Chemistry and Molecular Bioscience, Molecular Horizons, and Australian Research Council Centre for Cryo-electron Microscopy of Membrane Proteins, University of Wollongong, Wollongong, NSW, Australia
| | - Peter D. Mace
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
5
|
Merino-Cacho L, Barroso-Gomila O, Pozo-Rodríguez M, Muratore V, Guinea-Pérez C, Serrano Á, Pérez C, Cano-López S, Urcullu A, Azkargorta M, Iloro I, Galdeano C, Juárez-Jiménez J, Mayor U, Elortza F, Barrio R, Sutherland JD. Cullin-RING ligase BioE3 reveals molecular-glue-induced neosubstrates and rewiring of the endogenous Cereblon ubiquitome. Cell Commun Signal 2025; 23:101. [PMID: 39972349 PMCID: PMC11841277 DOI: 10.1186/s12964-025-02091-5] [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: 10/25/2024] [Accepted: 02/08/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The specificity of the ubiquitination process is mediated by the E3 ligases. Discriminating genuine substrates of E3s from mere interacting proteins is one of the major challenges in the field. We previously developed BioE3, a biotin-based approach that uses BirA-E3 fusions together with ubiquitin fused to a low-affinity AviTag to obtain a site-specific and proximity-dependent biotinylation of the substrates. We proved the suitability of BioE3 to identify targets of RING and HECT-type E3 ligases. METHODS BioE3 experiments were performed in HEK293FT and U2OS stable cell lines expressing TRIPZ-bioGEFUb transiently transfected with BirA-cereblon (CRBN). Cells were seeded using biotin-free media, followed later by a short-biotin pulse. We evaluated the applicability of the BioE3 system to CRBN and molecular glues by Western blot and confocal microscopy, blocking the proteasome with bortezomib, inhibiting NEDDylation with MLN4924 and treating the cells with pomalidomide. For the identification of endogenous substrates and neosubstrates we analyzed the eluates of streptavidin pull-downs of BioE3 experiments by LC-MS/MS. Analysis of targets for which ubiquitination changes significantly upon treatment was done using two-sided Student's t-test. Orthogonal validations were performed by histidine pull-down, GFP-trap and computational modelling. RESULTS Here we demonstrate that BioE3 is suitable for the multi-protein complex Cullin-RING E3s ligases (CRLs), the most utilized E3-type for targeted protein degradation (TPD) strategies. Using CRBN as proof of concept, one of the substrate receptors of CRL4 E3 ligase, we identified both endogenous substrates and novel neosubstrates upon pomalidomide treatment, including CSDE1 which contains a G-loop motif potentially involved in the binding to CRBN in presence of pomalidomide. Importantly, we observed a major rearrangement of the endogenous ubiquitination landscape upon treatment with this molecular glue. CONCLUSIONS The ability of BioE3 to detect and compare both substrates and neosubstrates, as well as how substrates change in response to treatments, will facilitate both on-target and off-target identifications and offer a broader characterization and validation of TPD compounds, like molecular glues and PROTACs.
Collapse
Affiliation(s)
- Laura Merino-Cacho
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
| | - Orhi Barroso-Gomila
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
- Present address: Biobizkaia Health Research Institute, Barakaldo, 48903, Spain
| | - Mónica Pozo-Rodríguez
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
| | - Veronica Muratore
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
- Present address: Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Claudia Guinea-Pérez
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
| | - Álvaro Serrano
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy, University of Barcelona, 08028, Barcelona, Spain
- Institute of Theoretical and Computational Chemistry (IQTC), School of Chemistry and Physics, University of Barcelona, 08028, Barcelona, Spain
| | - Coralia Pérez
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
| | - Sandra Cano-López
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
| | - Ainhoa Urcullu
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
- Instituto de Salud Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Ibon Iloro
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
- Instituto de Salud Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Carles Galdeano
- Institute of Theoretical and Computational Chemistry (IQTC), School of Chemistry and Physics, University of Barcelona, 08028, Barcelona, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, 08028, Barcelona, Spain
| | - Jordi Juárez-Jiménez
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy, University of Barcelona, 08028, Barcelona, Spain
- Institute of Theoretical and Computational Chemistry (IQTC), School of Chemistry and Physics, University of Barcelona, 08028, Barcelona, Spain
| | - Ugo Mayor
- Biochemistry and Molecular Biology Department, University of the Basque Country, Leioa, Spain
- Ikerbasque-Basque Foundation for Science, Bilbao, Spain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain
- Instituto de Salud Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain.
| | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC Biogune), Basque Research and Technology Alliance (BRTA), 48160, Derio, Spain.
| |
Collapse
|
6
|
Karayel O, Soung A, Gurung H, Schubert AF, Klaeger S, Kschonsak M, Al-Maraghi A, Bhat AA, Alshabeeb Akil AS, Dugger DL, Webster JD, French DM, Anand D, Soni N, Fakhro KA, Rose CM, Harris SF, Ndoja A, Newton K, Dixit VM. Impairment of DET1 causes neurological defects and lethality in mice and humans. Proc Natl Acad Sci U S A 2025; 122:e2422631122. [PMID: 39937864 PMCID: PMC11848315 DOI: 10.1073/pnas.2422631122] [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: 10/31/2024] [Accepted: 01/14/2025] [Indexed: 02/14/2025] Open
Abstract
COP1 and DET1 are components of an E3 ubiquitin ligase that is conserved from plants to humans. Mammalian COP1 binds to DET1 and is a substrate adaptor for the CUL4A-DDB1-RBX1 RING E3 ligase. Transcription factor substrates, including c-Jun, ETV4, and ETV5, are targeted for proteasomal degradation to effect rapid transcriptional changes in response to cues such as growth factor deprivation. Here, we link a homozygous DET1R26W mutation to lethal developmental abnormalities in humans. Experimental cryo-electron microscopy of the DET1 complex with DDB1 and DDA1, as well as co-immunoprecipitation experiments, revealed that DET1R26W impairs binding to DDB1, thereby compromising E3 ligase function. Accordingly, human-induced pluripotent stem cells homozygous for DET1R26W expressed ETV4 and ETV5 highly, and exhibited defective mitochondrial homeostasis and aberrant caspase-dependent cell death when differentiated into neurons. Neuronal cell death was increased further in the presence of Det1-deficient microglia as compared to WT microglia, indicating that the deleterious effects of the DET1 p.R26W mutation may stem from the dysregulation of multiple cell types. Mice lacking Det1 died during embryogenesis, while Det1 deletion just in neural stem cells elicited hydrocephalus, cerebellar dysplasia, and neonatal lethality. Our findings highlight an important role for DET1 in the neurological development of mice and humans.
Collapse
Affiliation(s)
- Ozge Karayel
- Department of Physiological Chemistry, Genentech, South San Francisco, CA94080
| | - Allison Soung
- Department of Neuroscience, Genentech, South San Francisco, CA94080
| | - Hem Gurung
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA94080
| | | | - Susan Klaeger
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA94080
| | - Marc Kschonsak
- Department of Structural Biology, Genentech, South San Francisco, CA94080
| | | | - Ajaz A. Bhat
- Department of Human Genetics, Sidra Medicine, Doha26999, Qatar
| | | | - Debra L. Dugger
- Department of Physiological Chemistry, Genentech, South San Francisco, CA94080
| | | | | | | | - Naharmal Soni
- Division of Neonatology, Sidra Medicine, Doha26999, Qatar
| | - Khalid A. Fakhro
- Department of Human Genetics, Sidra Medicine, Doha26999, Qatar
- Genomics, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha34110, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha24144, Qatar
| | - Christopher M. Rose
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA94080
| | - Seth F. Harris
- Department of Structural Biology, Genentech, South San Francisco, CA94080
| | - Ada Ndoja
- Department of Neuroscience, Genentech, South San Francisco, CA94080
| | - Kim Newton
- Department of Physiological Chemistry, Genentech, South San Francisco, CA94080
| | - Vishva M. Dixit
- Department of Physiological Chemistry, Genentech, South San Francisco, CA94080
| |
Collapse
|
7
|
Guo R, Yang F, Cherney EC. Small molecule targeted protein degradation via the UPS: venturing beyond E3 substrate receptors. RSC Med Chem 2025:d4md00718b. [PMID: 39949641 PMCID: PMC11815867 DOI: 10.1039/d4md00718b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/10/2024] [Indexed: 02/16/2025] Open
Abstract
The ubiquitin proteasome system (UPS) has been successfully hi-jacked by both bifunctional and monovalent small molecules to affect the degradation of proteins that were once considered undruggable. This field has primarily focused on the targeted recruitment of proteins to substrate receptors on E3 ubiquitin ligases, which are only one part of the UPS. More recently, the field has begun to explore recruitment to other types of UPS proteins including E2 ubiquitin-conjugating enzymes, substrate adaptor proteins within the E3 complex, chaperone proteins that associate with E3s, proteasomal subunits, and proteasome-associated proteins. While these approaches are relatively nascent compared to more traditional E3 substrate receptor-based degradation, these approaches are starting to show promise and could offer unique advantages. This review will cover key findings in small molecule UPS-mediated targeted protein degradation (TPD) affected by co-opting proteins beyond traditional E3 substrate receptors.
Collapse
Affiliation(s)
- Renyu Guo
- Discovery & Development Sciences, Bristol Myers Squibb Co Princeton New Jersey 08543 USA
| | - Fukang Yang
- Discovery & Development Sciences, Bristol Myers Squibb Co Princeton New Jersey 08543 USA
| | - Emily C Cherney
- Discovery & Development Sciences, Bristol Myers Squibb Co Princeton New Jersey 08543 USA
| |
Collapse
|
8
|
Rodrigues da Silva EV, Torres C, Nemamiah Escolarique Ribeiro H, Santana Travaglini Berti de Correia CR, de Oliveira de Castro T, da Costa Mancin G, Zanchetta Venancio MG, Abdel Baqui MM, Teixeira FR, Gomes MD. Molecular characterization of the E2 conjugating enzyme LinfUbc13 in Leishmania infantum. Arch Biochem Biophys 2025; 764:110272. [PMID: 39689751 DOI: 10.1016/j.abb.2024.110272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
UBC13 is an orthologue of Homo sapiens ubiquitin-conjugation E2 enzymes described in Leishmania mexicana, a null mutant lacking this gene cannot be produced, suggesting essential functions in this parasite. Leishmania infantum is an etiological agent of visceral leishmaniasis, the most severe type of disease that is potentially fatal if untreated. The ubiquitination process has been targeted for leishmanicidal compounds, indicating its essential function in parasite homeostasis. Therefore, the molecular characterization of the ubiquitination process may provide a better understanding of the molecular and cellular basis of leishmaniasis. Here, we characterized the gene LINF_350017900 in Leishmania infantum, which was named LinfUBC13, an E2 orthologue of UBC13 in Leishmania mexicana and the UBE2D family in Homo sapiens, sharing 72-74 % identity with UBE2D1, UBE2D2, and UBE2D3. LinfUbc13 contains conserved catalytic residues, including Cys86 and the HPN motif, which are essential for ubiquitin-conjugating activity. Structural analysis revealed a high similarity between LinfUbc13 and human UBE2D proteins, with a root-mean-square deviation (RMSD) of 0.4 Å, suggesting conserved functions. Recombinant LinfUbc13 was expressed and shown to accept ubiquitin from E1, forming a thioester intermediate. Functional assays demonstrated that LinfUbc13 transfers ubiquitin to p53 through human HDM2 E3 ligase, confirming its role in ubiquitination. Subcellular localization showed that LinfUbc13 was distributed throughout the parasite cytoplasm. These findings highlight the conserved nature of the ubiquitin-proteasome system between Leishmania infantum and Homo sapiens, showing that LinfUbc13 is an E2 enzyme that plays a crucial role in parasitic development.
Collapse
Affiliation(s)
| | - Caroline Torres
- Department of Genetics and Evolution, Federal University of São Carlos, São Carlos, Brazil; Current address: Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Camila Rolemberg Santana Travaglini Berti de Correia
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Genetics and Evolution, Federal University of São Carlos, São Carlos, Brazil
| | | | - Giovanna da Costa Mancin
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mayla Gabriela Zanchetta Venancio
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical Scool, University of São Paulo, Ribeirão Preto, Brazil
| | - Munira Muhammad Abdel Baqui
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical Scool, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Roberti Teixeira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Genetics and Evolution, Federal University of São Carlos, São Carlos, Brazil; Current address: Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| | - Marcelo Damário Gomes
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
9
|
Rosignoli S, Giordani S, Pacelli M, Guarguaglini G, Paiardini A. Unraveling the Engagement of Kinases to CRBN Through a Shared Structural Motif to Optimize PROTACs Efficacy. Biomolecules 2025; 15:206. [PMID: 40001507 PMCID: PMC11852972 DOI: 10.3390/biom15020206] [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: 12/31/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
PROteolysis TArgeting Chimeras (PROTACs) offer a therapeutic modality for protein target engagement, exploiting the ubiquitin-proteasome system to achieve precise degradation of a protein of interest. Recent advancements in understanding the structural biology of the CRL4A E3 ligase complex, particularly its recruitment of neo-substrates through the G-loop motif, have provided valuable insights into the optimization of PROTAC efficacy. This perspective delves into the molecular determinants governing PROTAC selectivity and degradation efficiency, with a specific focus on kinases showing distinct G-loop conformations. By employing computational approaches to predict ternary complexes, along with the identification of binding patterns, it is possible to address limitations posed by structural data scarcity, thereby enhancing rational design strategies.
Collapse
Affiliation(s)
- Serena Rosignoli
- Centre for Regenerative Medicine “Stefano Ferrari”, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Sara Giordani
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (S.G.); (M.P.)
| | - Maddalena Pacelli
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (S.G.); (M.P.)
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, 00185 Rome, Italy;
| | - Alessandro Paiardini
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (S.G.); (M.P.)
| |
Collapse
|
10
|
Baker HA, Bernardini JP, Csizmók V, Madero A, Kamat S, Eng H, Lacoste J, Yeung FA, Comyn S, Hui E, Calabrese G, Raught B, Taipale M, Mayor T. The co-chaperone DNAJA2 buffers proteasomal degradation of cytosolic proteins with missense mutations. J Cell Sci 2025; 138:jcs262019. [PMID: 39618332 DOI: 10.1242/jcs.262019] [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: 02/12/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
Mutations can disrupt the native function of protein by causing misfolding, which is generally handled by an intricate protein quality control network. To better understand the triaging mechanisms for misfolded cytosolic proteins, we screened a human mutation library to identify a panel of unstable mutations. The degradation of these mutated cytosolic proteins is largely dependent on the ubiquitin proteasome system. Using BioID proximity labelling, we found that the co-chaperones DNAJA1 and DNAJA2 are key interactors with one of the mutated proteins. Notably, the absence of DNAJA2 increases the turnover of the mutant but not the wild-type protein. Our work indicates that specific missense mutations in cytosolic proteins can promote enhanced interactions with molecular chaperones. Assessment of the broader panel of cytosolic mutant proteins shows that the co-chaperone DNAJA2 exhibits two distinct behaviours - acting to stabilize a wide array of cytosolic proteins, including wild-type variants, and to specifically 'buffer' some mutant proteins to reduce their turnover. Our work illustrates how distinct elements of the protein homeostasis network are utilized in the presence of a cytosolic misfolded protein.
Collapse
Affiliation(s)
- Heather A Baker
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Edwin SH Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Jonathan P Bernardini
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Veronika Csizmók
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Angel Madero
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Shriya Kamat
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Edwin SH Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Hailey Eng
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Jessica Lacoste
- Department of Molecular Genetics, Terrence Donnelly Centre for Cellular & Biomedical Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Faith A Yeung
- Princess Margaret Cancer Centre, University Health Network and Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Sophie Comyn
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Elizabeth Hui
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Gaetano Calabrese
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network and Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Mikko Taipale
- Department of Molecular Genetics, Terrence Donnelly Centre for Cellular & Biomedical Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Thibault Mayor
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Edwin SH Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
11
|
Cornelius RJ, Maeoka Y, Shinde U, McCormick JA. Familial Hyperkalemic Hypertension. Compr Physiol 2024; 14:5839-5874. [PMID: 39699086 DOI: 10.1002/cphy.c240004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
The rare disease Familial Hyperkalemic Hypertension (FHHt) is caused by mutations in the genes encoding Cullin 3 (CUL3), Kelch-Like 3 (KLHL3), and two members of the With-No-Lysine [K] (WNK) kinase family, WNK1 and WNK4. In the kidney, these mutations ultimately cause hyperactivation of NCC along the renal distal convoluted tubule. Hypertension results from increased NaCl retention, and hyperkalemia by impaired K + secretion by downstream nephron segments. CUL3 and KLHL3 are now known to form a ubiquitin ligase complex that promotes proteasomal degradation of WNK kinases, which activate downstream kinases that phosphorylate and thus activate NCC. For CUL3, potent effects on the vasculature that contribute to the more severe hypertensive phenotype have also been identified. Here we outline the in vitro and in vivo studies that led to the discovery of the molecular pathways regulating NCC and vascular tone, and how FHHt-causing mutations disrupt these pathways. Potential mechanisms for variability in disease severity related to differential effects of each mutation on the kidney and vasculature are described, and other possible effects of the mutant proteins beyond the kidney and vasculature are explored. © 2024 American Physiological Society. Compr Physiol 14:5839-5874, 2024.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Yujiro Maeoka
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Ujwal Shinde
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
12
|
Kienle SM, Schneider T, Bernecker C, Bracker J, Marx A, Kovermann M, Scheffner M, Stuber K. Biochemical and Structural Consequences of NEDD8 Acetylation. Chembiochem 2024; 25:e202400478. [PMID: 39022855 DOI: 10.1002/cbic.202400478] [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: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/20/2024]
Abstract
Similar to ubiquitin, the ubiquitin-like protein NEDD8 is not only conjugated to other proteins but is itself subject to posttranslational modifications including lysine acetylation. Yet, compared to ubiquitin, only little is known about the biochemical and structural consequences of site-specific NEDD8 acetylation. Here, we generated site-specifically mono-acetylated NEDD8 variants for each known acetylation site by genetic code expansion. We show that, in particular, acetylation of K11 has a negative impact on the usage of NEDD8 by the NEDD8-conjugating enzymes UBE2M and UBE2F and that this is likely due to electrostatic and steric effects resulting in conformational changes of NEDD8. Finally, we provide evidence that p300 acts as a position-specific NEDD8 acetyltransferase.
Collapse
Affiliation(s)
- Simon Maria Kienle
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Tobias Schneider
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Christine Bernecker
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Janina Bracker
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Andreas Marx
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Michael Kovermann
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Martin Scheffner
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Katrin Stuber
- Departments of Biology and Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| |
Collapse
|
13
|
Jiang Y, Le F, Huang S, Chen X, Deng Z. MLN4924 Suppresses head and neck squamous cell carcinoma progression by inactivating the mTOR signaling pathway via the NEDD8/CUL4/TSC2 axis. Int J Biochem Cell Biol 2024; 177:106696. [PMID: 39566655 DOI: 10.1016/j.biocel.2024.106696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an aggressive cancer with a five-year survival rate below 50 %. Standard treatments for HNSCC include surgery, radiotherapy, chemotherapy, and targeted therapies, but they still have significant limitations. Neddylation, a post-translational modification involving the attachment of NEDD8 (neural precursor cells expressed developmentally down-regulated 8) to proteins, is frequently dysregulated in HNSCC, thereby promoting tumor growth. MLN4924, also known as Pevonedistat, is a Neddylation inhibitor that has shown promise in suppressing HNSCC cell proliferation and invasion, establishing it as a potential therapeutic option. However, its precise molecular mechanism remains unclear. This study aims to investigate the mechanism of MLN4924 in HNSCC. This study examined the effects of MLN4924 on HNSCC and its associated molecular pathways. Bioinformatic analysis indicated that NEDD8, a critical component of the Neddylation pathway, is linked to poor prognosis and the mTOR (mammalian target of rapamycin) signaling pathway in HNSCC. MLN4924 significantly suppressed cell migration, invasion, and the epithelial-mesenchymal transition (EMT) pathway, and downregulated NEDD8 expression. Mechanistic studies demonstrated that MLN4924 inhibited the binding of NEDD8 to cullin4 (CUL4) and prevented the Neddylation of tuberous sclerosis complex 2 (TSC2), leading to the inactivation of the mTOR pathway. These findings were confirmed in vivo, where MLN4924 effectively inhibited tumor growth. Overall, MLN4924 disrupted Neddylation pathway and stabilized TSC2, thereby inactivating the mTOR pathway. The study provided a theoretical basis for the clinical potential of MLN4924 in improving treatment outcomes for HNSCC patients, offering a novel strategy for addressing this challenging disease.
Collapse
Affiliation(s)
- Youfang Jiang
- Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, China; Department of head and neck Surgery, Jiangxi Cancer hospital, Nanchang Medical College, Nanchang, Jiangxi 330029, China
| | - Fei Le
- Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, China; Department of head and neck Surgery, Jiangxi Cancer hospital, Nanchang Medical College, Nanchang, Jiangxi 330029, China
| | - Shuangling Huang
- Department of neurosurgery, Jiangxi Cancer hospital, Nanchang Medical College, Nanchang, Jiangxi 330029, China
| | - Xuezhong Chen
- Department of Nuclear Medicine, First affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China
| | - Ziqing Deng
- Department of General Surgery, The Third Hospital of Nanchang, Nanchang, Jiangxi 330000, China.
| |
Collapse
|
14
|
Scott DC, Chittori S, Purser N, King MT, Maiwald SA, Churion K, Nourse A, Lee C, Paulo JA, Miller DJ, Elledge SJ, Harper JW, Kleiger G, Schulman BA. Structural basis for C-degron selectivity across KLHDCX family E3 ubiquitin ligases. Nat Commun 2024; 15:9899. [PMID: 39548056 PMCID: PMC11568203 DOI: 10.1038/s41467-024-54126-z] [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: 08/20/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Specificity of the ubiquitin-proteasome system depends on E3 ligase-substrate interactions. Many such pairings depend on E3 ligases binding to peptide-like sequences - termed N- or C-degrons - at the termini of substrates. However, our knowledge of structural features distinguishing closely related C-degron substrate-E3 pairings is limited. Here, by systematically comparing ubiquitylation activities towards a suite of common model substrates, and defining interactions by biochemistry, crystallography, and cryo-EM, we reveal principles of C-degron recognition across the KLHDCX family of Cullin-RING ligases (CRLs). First, a motif common across these E3 ligases anchors a substrate's C-terminus. However, distinct locations of this C-terminus anchor motif in different blades of the KLHDC2, KLHDC3, and KLHDC10 β-propellers establishes distinct relative positioning and molecular environments for substrate C-termini. Second, our structural data show KLHDC3 has a pre-formed pocket establishing preference for an Arg or Gln preceding a C-terminal Gly, whereas conformational malleability contributes to KLHDC10's recognition of varying features adjacent to substrate C-termini. Finally, additional non-consensus interactions, mediated by C-degron binding grooves and/or by distal propeller surfaces and substrate globular domains, can substantially impact substrate binding and ubiquitylatability. Overall, the data reveal combinatorial mechanisms determining specificity and plasticity of substrate recognition by KLDCX-family C-degron E3 ligases.
Collapse
Affiliation(s)
- Daniel C Scott
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sagar Chittori
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nicholas Purser
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Moeko T King
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Samuel A Maiwald
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Kelly Churion
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amanda Nourse
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chan Lee
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen J Elledge
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Gary Kleiger
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Brenda A Schulman
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
15
|
Winter GE. Extrapolating Lessons from Targeted Protein Degradation to Other Proximity-Inducing Drugs. ACS Chem Biol 2024; 19:2089-2102. [PMID: 39264973 PMCID: PMC11494510 DOI: 10.1021/acschembio.4c00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024]
Abstract
Targeted protein degradation (TPD) is an emerging pharmacologic strategy. It relies on small-molecule "degraders" that induce proximity of a component of an E3 ubiquitin ligase complex and a target protein to induce target ubiquitination and subsequent proteasomal degradation. Essentially, degraders thus expand the function of E3 ligases, allowing them to degrade proteins they would not recognize in the absence of the small molecule. Over the past decade, insights gained from identifying, designing, and characterizing various degraders have significantly enhanced our understanding of TPD mechanisms, precipitating in rational degrader discovery strategies. In this Account, I aim to explore how these insights can be extrapolated to anticipate both opportunities and challenges of utilizing the overarching concept of proximity-inducing pharmacology to manipulate other cellular circuits for the dissection of biological mechanisms and for therapeutic purposes.
Collapse
Affiliation(s)
- Georg E. Winter
- CeMM Research Center for
Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| |
Collapse
|
16
|
Zhong G, Fang Z, Sun T, Ying M, Wang A, Chen Y, Wang H, Ma C, Wang C, Ge R, Liu G, Guo Y. Ubiquitin ligase RFWD2 promotes dendritic spine and synapse formation by activating the ERK/PEA3/c-Jun pathway in rat cerebral cortical neurons. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167319. [PMID: 38909848 DOI: 10.1016/j.bbadis.2024.167319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
The regulation of protein degradation through the ubiquitin-proteasome system is essential for normal brain development, axon growth, synaptic growth and plasticity. The E3 ubiquitin ligase RFWD2 plays a key role in the onset and development of neurological diseases, including the pathogenesis of Alzheimer's disease (AD), but the mechanisms controlling the homeostasis of neuronal synaptic proteins are still poorly understood. Here, we showed that the expression level of RFWD2 gradually decreased with the age of the rats and was negatively correlated with the development of cerebral cortical neurons and dendrites in vivo. RFWD2 was shown to localize to presynaptic terminals and some postsynaptic sides of both excitatory synapses and inhibitory synapses via colocalization with neuronal synaptic proteins (SYN, PSD95, Vglut1 and GAD67). Overexpression of RFWD2 promoted dendrite development and dendritic spine formation and markedly decreased the expression of synaptophysin and PSD95 by reducing the expression of ETV1, ETV4, ETV5 and c-JUN in vitro. Furthermore, the whole-cell membrane slice clamp results showed that RFWD2 overexpression resulted in greater membrane capacitance in neuronal cells, inadequate cell repolarization, and a longer time course for neurons to emit action potentials with decreased excitability. RFWD2 regulates dendritic development and plasticity, dendritic spine formation and synaptic function in rat cerebral cortex neurons by activating the ERK/PEA3/c-Jun pathway via a posttranslational regulatory mechanism and can be used as an efficient treatment target for neurological diseases.
Collapse
Affiliation(s)
- Guangshang Zhong
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Zhuling Fang
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Tingting Sun
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Mengjiao Ying
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Ao Wang
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Ying Chen
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Haojie Wang
- School of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Caiyun Ma
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Chunjing Wang
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Rongjing Ge
- School of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, 233000, China
| | - Gaofeng Liu
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China; Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, 233000, China.
| | - Yu Guo
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, 233000, China.
| |
Collapse
|
17
|
Qin X, Han X, Sun Y. Discovery of small molecule inhibitors of neddylation catalyzing enzymes for anticancer therapy. Biomed Pharmacother 2024; 179:117356. [PMID: 39214012 DOI: 10.1016/j.biopha.2024.117356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Protein neddylation, a type of post-translational modifications, involves the transfer of the ubiquitin-like protein NEDD8 to the lysine residues of a target substrate, which is catalyzed by the NEDD8 activating enzyme (E1), NEDD8 conjugating enzyme (E2), and NEDD8 ligase (E3). Cullin family proteins, core components of Cullin-RING E3 ubiquitin ligases (CRLs), are the most well-known physiological substrates of neddylation. CRLs, activated upon cullin neddylation, promote the ubiquitination of a variety of key signaling proteins for proteasome degradation, thereby regulating many critical biological functions. Abnormal activation of neddylation enzymes as well as CRLs has been frequently observed in various human cancers and is associated with poor prognosis for cancer patients. Consequently, targeting neddylation has emerged as a promising strategy for the development of novel anticancer therapeutics. This review first briefly introduces the properties of protein neddylation and its role in cancer, and then systematically summarizes all reported chemical inhibitors of the three neddylation enzymes, providing a focused, up to date, and comprehensive resource in the discovery and development of these small molecule inhibitors.
Collapse
Affiliation(s)
- Xiangshuo Qin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Cancer Center of Zhejiang University, Hangzhou 310029, China
| | - Xin Han
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Cancer Center of Zhejiang University, Hangzhou 310029, China.
| | - Yi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Cancer Center of Zhejiang University, Hangzhou 310029, China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| |
Collapse
|
18
|
Kwon DH, Shin S, Nam YS, Choe N, Lim Y, Jeong A, Lee YG, Kim YK, Kook H. CBL-b E3 ligase-mediated neddylation and activation of PARP-1 induce vascular calcification. Exp Mol Med 2024; 56:2246-2259. [PMID: 39349831 PMCID: PMC11541702 DOI: 10.1038/s12276-024-01322-y] [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: 02/08/2024] [Revised: 06/24/2024] [Accepted: 07/16/2024] [Indexed: 10/03/2024] Open
Abstract
Vascular calcification (VC) refers to the accumulation of mineral deposits on the walls of arteries and veins, and it is closely associated with increased mortality in cardiovascular disease patients, particularly among high-risk patients with diabetes and chronic kidney disease (CKD). Neuronal precursor cell-expressed developmentally downregulated protein 8 (NEDD8) is a ubiquitin-like protein that plays a pivotal role in various cellular functions, primarily through its conjugation to target proteins and subsequent relay of biological signals. However, the role of NEDDylation in VC has not been investigated. In our study, we observed that MLN4924, an inhibitor of the NEDD8-activating E1 enzyme, effectively impedes the progression of VC. LC‒MS/MS analysis revealed that poly(ADP‒ribose) polymerase 1 (PARP-1) is subjected to NEDD8 conjugation, leading to an increase in PARP-1 activity during VC. We subsequently revealed that PARP-1 NEDDylation is mediated by the E3 ligase CBL proto-oncogene B (CBL-b) and is reversed by NEDD8-specific protease 1 (NEDP-1) during VC. Furthermore, the CBL-b C373 peptide effectively mitigated the inactive form of the E3 ligase activity of CBL-b, ultimately preventing VC. These findings provide compelling evidence that the NEDD8-dependent activation of PARP-1 represents a novel mechanism underlying vascular calcification and suggests a promising new therapeutic target for VC.
Collapse
Affiliation(s)
- Duk-Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea.
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea.
- BK21 plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, Republic of Korea.
| | - Sera Shin
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Yoon Seok Nam
- Institute of Well-Aging Medicare & CSU G-LAMP Project Group, Chosun University, Gwangju, Republic of Korea
| | - Nakwon Choe
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Yongwoon Lim
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Anna Jeong
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
- BK21 plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, Republic of Korea
| | - Yun-Gyeong Lee
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Young-Kook Kim
- BK21 plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, Republic of Korea
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea.
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea.
- BK21 plus Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
19
|
Bou Malhab LJ, Schmidt S, Fagotto-Kaufmann C, Pion E, Gadea G, Roux P, Fagotto F, Debant A, Xirodimas DP. An Anti-Invasive Role for Mdmx through the RhoA GTPase under the Control of the NEDD8 Pathway. Cells 2024; 13:1625. [PMID: 39404389 PMCID: PMC11475522 DOI: 10.3390/cells13191625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Mdmx (Mdm4) is established as an oncogene mainly through repression of the p53 tumour suppressor. On the other hand, anti-oncogenic functions for Mdmx have also been proposed, but the underlying regulatory pathways remain unknown. Investigations into the effect of inhibitors for the NEDD8 pathway in p53 activation, human cell morphology, and in cell motility during gastrulation in Xenopus embryos revealed an anti-invasive function of Mdmx. Through stabilisation and activation of the RhoA GTPase, Mdmx is required for the anti-invasive effects of NEDDylation inhibitors. Mechanistically, through its Zn finger domain, Mdmx preferentially interacts with the inactive GDP-form of RhoA. This protects RhoA from degradation and allows for RhoA targeting to the plasma membrane for its subsequent activation. The effect is transient, as prolonged NEDDylation inhibition targets Mdmx for degradation, which subsequently leads to RhoA destabilisation. Surprisingly, Mdmx degradation requires non-NEDDylated (inactive) Culin4A and the Mdm2 E3-ligase. This study reveals that Mdmx can control cell invasion through RhoA stabilisation/activation, which is potentially linked to the reported anti-oncogenic functions of Mdmx. As inhibitors of the NEDD8 pathway are in clinical trials, the status of Mdmx may be a critical determinant for the anti-tumour effects of these inhibitors.
Collapse
Affiliation(s)
- Lara J. Bou Malhab
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Susanne Schmidt
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Christine Fagotto-Kaufmann
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Emmanuelle Pion
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Gilles Gadea
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Pierre Roux
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Francois Fagotto
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Anne Debant
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Dimitris P. Xirodimas
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| |
Collapse
|
20
|
Blackburn PR, Ebstein F, Hsieh TC, Motta M, Radio FC, Herkert JC, Rinne T, Thiffault I, Rapp M, Alders M, Maas S, Gerard B, Smol T, Vincent-Delorme C, Cogné B, Isidor B, Vincent M, Bachmann-Gagescu R, Rauch A, Joset P, Ferrero GB, Ciolfi A, Husson T, Guerrot AM, Bacino C, Macmurdo C, Thompson SS, Rosenfeld JA, Faivre L, Mau-Them FT, Deb W, Vignard V, Agrawal PB, Madden JA, Goldenberg A, Lecoquierre F, Zech M, Prokisch H, Necpál J, Jech R, Winkelmann J, Koprušáková MT, Konstantopoulou V, Younce JR, Shinawi M, Mighton C, Fung C, Morel CF, Lerner-Ellis J, DiTroia S, Barth M, Bonneau D, Krapels I, Stegmann APA, van der Schoot V, Brunet T, Bußmann C, Mignot C, Zampino G, Wortmann SB, Mayr JA, Feichtinger RG, Courtin T, Ravelli C, Keren B, Ziegler A, Hasadsri L, Pichurin PN, Klee EW, Grand K, Sanchez-Lara PA, Krüger E, Bézieau S, Klinkhammer H, Krawitz PM, Eichler EE, Tartaglia M, Küry S, Wang T. Loss-of-Function Variants in CUL3 Cause a Syndromic Neurodevelopmental Disorder. Ann Neurol 2024; 97:76-89. [PMID: 39301775 PMCID: PMC11922793 DOI: 10.1002/ana.27077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE De novo variants in cullin-3 ubiquitin ligase (CUL3) have been strongly associated with neurodevelopmental disorders (NDDs), but no large case series have been reported so far. Here, we aimed to collect sporadic cases carrying rare variants in CUL3, describe the genotype-phenotype correlation, and investigate the underlying pathogenic mechanism. METHODS Genetic data and detailed clinical records were collected via multicenter collaboration. Dysmorphic facial features were analyzed using GestaltMatcher. Variant effects on CUL3 protein stability were assessed using patient-derived T-cells. RESULTS We assembled a cohort of 37 individuals with heterozygous CUL3 variants presenting a syndromic NDD characterized by intellectual disability with or without autistic features. Of these, 35 have loss-of-function (LoF) and 2 have missense variants. CUL3 LoF variants in patients may affect protein stability leading to perturbations in protein homeostasis, as evidenced by decreased ubiquitin-protein conjugates in vitro. Notably, we show that 4E-BP1 (EIF4EBP1), a prominent substrate of CUL3, fails to be targeted for proteasomal degradation in patient-derived cells. INTERPRETATION Our study further refines the clinical and mutational spectrum of CUL3-associated NDDs, expands the spectrum of cullin RING E3 ligase-associated neuropsychiatric disorders, and suggests haploinsufficiency via LoF variants is the predominant pathogenic mechanism. ANN NEUROL 2024.
Collapse
Affiliation(s)
- Patrick R Blackburn
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Frédéric Ebstein
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
| | - Tzung-Chien Hsieh
- Institute of Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
| | - Marialetizia Motta
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | - Johanna C Herkert
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tuula Rinne
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Isabelle Thiffault
- Center for Pediatric Genomic Medicine, Children's Mercy Hospital, Kansas City, MO, USA
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospitals, Kansas City, MO, USA
| | - Michele Rapp
- Department of Pediatrics-Clinical Genetics and Metabolism, Children's Hospital Colorado, Aurora, CO, USA
| | - Mariel Alders
- Amsterdam University Medical Center, University of Amsterdam, Department of Clinical Genetics, Amsterdam, The Netherlands
| | - Saskia Maas
- Amsterdam University Medical Center, University of Amsterdam, Department of Clinical Genetics, Amsterdam, The Netherlands
| | - Bénédicte Gerard
- Unité de Biologie et de Génétique Moléculaire, Center Hospitalier Universitaire de Strasbourg, Strasbourg, France
| | - Thomas Smol
- Univ Lille, CHU Lille, RADEME Team, Institut de Génétique Médicale, Lille, France
| | | | - Benjamin Cogné
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique Médicale, Nantes, France
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique Médicale, Nantes, France
| | - Marie Vincent
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique Médicale, Nantes, France
| | - Ruxandra Bachmann-Gagescu
- Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Pascal Joset
- Medical Genetics, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Giovanni Battista Ferrero
- Department of Clinical and Biological Sciences, San Luigi Gonzaga University Hospital, University of Torino, Turin, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Thomas Husson
- Department of Research, Center Hospitalier du Rouvray, Rouen, France
- Normandie Univ, UNIROUEN, Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Developmental Disorders, Rouen, France
| | - Anne-Marie Guerrot
- Normandie Univ, UNIROUEN, Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Developmental Disorders, Rouen, France
| | - Carlos Bacino
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Colleen Macmurdo
- Division of Medical Genetics, Department of Internal Medicine, Baylor Scott and White Medical Center, Temple, TX, USA
| | - Stephanie S Thompson
- Division of Medical Genetics, Department of Internal Medicine, Baylor Scott and White Medical Center, Temple, TX, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Baylor Genetics, Houston, TX, USA
| | - Laurence Faivre
- Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD CHU, Dijon, France
- INSERM UMR1231, équipe GAD, Université de Bourgogne-Franche Comté, Dijon, France
| | - Frederic Tran Mau-Them
- INSERM UMR1231, équipe GAD, Université de Bourgogne-Franche Comté, Dijon, France
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Wallid Deb
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique Médicale, Nantes, France
| | - Virginie Vignard
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique Médicale, Nantes, France
| | - Pankaj B Agrawal
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston, MA, USA
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL, USA
| | - Jill A Madden
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston, MA, USA
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL, USA
| | - Alice Goldenberg
- Normandie Univ, UNIROUEN, Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Developmental Disorders, Rouen, France
| | - François Lecoquierre
- Normandie Univ, UNIROUEN, Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Developmental Disorders, Rouen, France
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Holger Prokisch
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Ján Necpál
- Department of Neurology, Zvolen Hospital, Zvolen, Slovakia
- Department of Neurology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Robert Jech
- Department of Neurology, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum Muenchen, Neuherberg, Germany
- Neurogenetics, Technische Universitaet Muenchen, Munich, Germany
- Institute of Human Genetics, Klinikum rechts der Isar der TUM, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | | | - John R Younce
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marwan Shinawi
- Division of Genetics and Genomic Medicine, St. Louis Children's Hospital, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chloe Mighton
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada
- Genomics Health Services and Policy Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Charlotte Fung
- The Fred A. Litwin Family Centre in Genetic Medicine, University Health Network and Sinai Health System, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Chantal F Morel
- The Fred A. Litwin Family Centre in Genetic Medicine, University Health Network and Sinai Health System, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Jordan Lerner-Ellis
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
| | - Stephanie DiTroia
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Magalie Barth
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
- Mitovasc Unit, UMR CNRS 6015-INSERM 1083, Angers, France
| | - Dominique Bonneau
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
| | - Ingrid Krapels
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht UMC, Maastricht, The Netherlands
| | - Alexander P A Stegmann
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht UMC, Maastricht, The Netherlands
| | - Vyne van der Schoot
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht UMC, Maastricht, The Netherlands
| | - Theresa Brunet
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Dr. v. Hauner Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU-University of Munich, Munich, Germany
| | - Cornelia Bußmann
- Department of Neuropediatrics, ATOS Klinik Heidelberg, Heidelberg, Germany
| | - Cyril Mignot
- Département de Génétique, AP-HP-Sorbonne Université, Hôpital Trousseau & Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Saskia B Wortmann
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Johannes A Mayr
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - René G Feichtinger
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Thomas Courtin
- Center for Molecular and Chromosomal Genetics, AP-HP-Sorbonne University, Pitié-Salpêtrière Hospital, Paris, France
| | - Claudia Ravelli
- Department of Pediatric Neurology and Neurogenetic Referral Center, AP-HP-Sorbonne Université, Armand Trousseau Hospital, Paris, France
| | - Boris Keren
- Département de Génétique, AP-HP-Sorbonne Université, Hôpital Trousseau & Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Alban Ziegler
- Mitovasc Unit, UMR CNRS 6015-INSERM 1083, Angers, France
- Department of Biochemistry and Genetics, Angers University Hospital and UMR CNRS, Angers, France
| | - Linda Hasadsri
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Pavel N Pichurin
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Eric W Klee
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Katheryn Grand
- Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Pedro A Sanchez-Lara
- Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Stéphane Bézieau
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique Médicale, Nantes, France
| | - Hannah Klinkhammer
- Institute of Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
- Institute of Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Peter Michael Krawitz
- Institute of Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Sébastien Küry
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique Médicale, Nantes, France
| | - Tianyun Wang
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
- Autism Research Center, Peking University Health Science Center, Beijing, China
| |
Collapse
|
21
|
Taylor JD, Barrett N, Martinez Cuesta S, Cassidy K, Pachl F, Dodgson J, Patel R, Eriksson TM, Riley A, Burrell M, Bauer C, Rees DG, Cimbro R, Zhang AX, Minter RR, Hunt J, Legg S. Targeted protein degradation using chimeric human E2 ubiquitin-conjugating enzymes. Commun Biol 2024; 7:1179. [PMID: 39300128 DOI: 10.1038/s42003-024-06803-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Proteins can be targeted for degradation by engineering biomolecules that direct them to the eukaryotic ubiquitination machinery. For instance, the fusion of an E3 ubiquitin ligase to a suitable target binding domain creates a 'biological Proteolysis-Targeting Chimera' (bioPROTAC). Here we employ an analogous approach where the target protein is recruited directly to a human E2 ubiquitin-conjugating enzyme via an attached target binding domain. Through rational design and screening we develop E2 bioPROTACs that induce the degradation of the human intracellular proteins SHP2 and KRAS. Using global proteomics, we characterise the target-specific and wider effects of E2 vs. VHL-based fusions. Taking SHP2 as a model target, we also employ a route to bioPROTAC discovery based on protein display libraries, yielding a degrader with comparatively weak affinity capable of suppressing SHP2-mediated signalling.
Collapse
Affiliation(s)
- Jonathan D Taylor
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK.
| | - Nathalie Barrett
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Sergio Martinez Cuesta
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Katelyn Cassidy
- Protein Sciences, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, 02451, USA
| | - Fiona Pachl
- Protein Sciences, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, 02451, USA
| | - James Dodgson
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Radhika Patel
- Centre for Genomics Research, Dynamic Omics, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Tuula M Eriksson
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Aidan Riley
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Matthew Burrell
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Christin Bauer
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - D Gareth Rees
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Raffaello Cimbro
- Centre for Genomics Research, Dynamic Omics, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Andrew X Zhang
- Protein Sciences, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, 02451, USA
| | - Ralph R Minter
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - James Hunt
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK.
| | - Sandrine Legg
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| |
Collapse
|
22
|
Bukhari Z, Gu L, Nederstigt AE, Cope LJ, Bolhuis DL, Harvey K, Allen T, Hill S, Yang Y, Lawson G, Lu C, Tran T, Pineda L, Low L, Chiang A, Song J, Fong MV, Rangel VM, Chan WK, Kleiger G, Goldfarb D, Vierra CA, Brown NG, Harrison JS. Design of linked-domain protein inhibitors of UBE2D as tools to study cellular ubiquitination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.02.610852. [PMID: 39282319 PMCID: PMC11398408 DOI: 10.1101/2024.09.02.610852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Ubiquitin (Ub) is a post-translational modification that largely controls proteostasis through mechanisms spanning transcription, translation, and notably, protein degradation. Ub conjugation occurs through a hierarchical cascade of three enzyme classes (E1, E2, and E3s) involving >1000 proteins that regulate the ubiquitination of proteins. The E2 Ub-conjugating enzymes are the midpoint, yet their cellular roles remain under-characterized, partly due to a lack of inhibitors. For example, the cellular roles of the promiscuous E2 UBE2D/UBCH5 are not well described. Here, we develop a highly selective, multivalent, engineered protein inhibitor for the UBE2D family that simultaneously targets the RING- and backside-binding sites. In HeLa cells, these inhibitors phenocopy knockdown of UBE2D by reducing the IC50 to cisplatin and whole-cell proteomics reveal an increased abundance of ~20% of the identified proteins, consistent with reduced Ub degradation and proteotoxic stress. These precision tools will enable new studies probing UBE2D's central role in proteome management.
Collapse
Affiliation(s)
- Zara Bukhari
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Li Gu
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | | | - Logan J Cope
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Derek L Bolhuis
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Kim Harvey
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Tristan Allen
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Spencer Hill
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Yujie Yang
- Department of Pharmaceutics & Medicinal Chemistry, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA
| | - Guy Lawson
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Cai Lu
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Tommy Tran
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Leah Pineda
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Leanne Low
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Andrew Chiang
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Jason Song
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Michelle V Fong
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - Vanessa M Rangel
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| | - William K Chan
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Gary Kleiger
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Dennis Goldfarb
- Department of Cell Biology and Physiology, Institute for Informatics, Washington University, St. Louis, MO, USA
| | - Craig A Vierra
- Biological Sciences Department, University of the Pacific, Stockton, CA 95211, USA
| | - Nicholas G Brown
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Joseph S Harrison
- The University of the Pacific, Department of Chemistry, Stockton, CA, 95210, USA
| |
Collapse
|
23
|
Yan YH, Wei LL, Wu JW, Wei SQ, Jiang YY, Yu JL, Yang LL, Li GB. Discovering New Metallo-Deubiquitinase CSN5 Inhibitors by a Non-Catalytic Activity Assay Platform. J Med Chem 2024; 67:14649-14667. [PMID: 39129245 DOI: 10.1021/acs.jmedchem.4c01514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
COP9 signalosome catalytic subunit CSN5 plays a key role in tumorigenesis and tumor immunity, showing potential as an anticancer target. Currently, only a few CSN5 inhibitors have been reported, at least partially, due to the challenges in establishing assays for CSN5 deubiquitinase activity. Here, we present the establishment and validation of a simple and reliable non-catalytic activity assay platform for identifying CSN5 inhibitors utilizing a new fluorescent probe, CFP-1, that exhibits enhanced fluorescence and fluorescence polarization features upon binding to CSN5. By using this platform, we identified 2-aminothiazole-4-carboxylic acids as new CSN5 inhibitors, which inhibited CSN5 but slightly downregulated PD-L1 in cancer cells. Furthermore, through the integration of deep learning-enabled virtual screening, we discovered that shikonins are nanomolar CSN5 inhibitors, which can upregulate PD-L1 in HCT116 cells. The binding modes of these structurally distinct inhibitors with CSN5 were explored by using microsecond-scale molecular dynamics simulations and tryptophan quenching assays.
Collapse
Affiliation(s)
- Yu-Hang Yan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Liu-Liu Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jing-Wei Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Si-Qi Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ying-Ying Jiang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Jun-Lin Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling-Ling Yang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Guo-Bo Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
24
|
Bell LE, Bardelle C, Packer MJ, Kastl J, Holdgate GA, Davies G. Characterisation of high throughput screening outputs for small molecule degrader discovery. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100162. [PMID: 38797285 DOI: 10.1016/j.slasd.2024.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Targeted protein degradation is an important mechanism carried out by the cellular machinery, one that is gaining momentum as an exploitable strategy for the development of drug-like compounds. Molecules which are able to induce proximity between elusive therapeutic targets of interest and E3 ligases which subsequently leads to proteasomal degradation of the target are beginning to decrease the percentage of the human proteome described as undruggable. Therefore, having the ability to screen for, and understand the mechanism of, such molecules is becoming an increasingly attractive scientific focus. We have established a number of cascade experiments including cell-based assays and orthogonal triage steps to provide annotation to the selectivity and mechanism of action for compounds identified as putative degraders from a primary high throughput screen against a high value oncology target. We will describe our current position, using PROTACs as proof-of-concept, on the analysis of these novel outputs and highlight challenges encountered.
Collapse
Affiliation(s)
- Lillie E Bell
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-BioCenter 1, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030, Vienna, Austria
| | - Catherine Bardelle
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| | | | - Johanna Kastl
- Assay.Works GmbH, Am Biopark 11, Regensburg, Germany
| | - Geoffrey A Holdgate
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK.
| | - Gareth Davies
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| |
Collapse
|
25
|
Chen X, Raiff A, Li S, Guo Q, Zhang J, Zhou H, Timms RT, Yao X, Elledge SJ, Koren I, Zhang K, Xu C. Mechanism of Ψ-Pro/C-degron recognition by the CRL2 FEM1B ubiquitin ligase. Nat Commun 2024; 15:3558. [PMID: 38670995 PMCID: PMC11053023 DOI: 10.1038/s41467-024-47890-5] [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: 10/13/2023] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The E3 ligase-degron interaction determines the specificity of the ubiquitin‒proteasome system. We recently discovered that FEM1B, a substrate receptor of Cullin 2-RING ligase (CRL2), recognizes C-degrons containing a C-terminal proline. By solving several cryo-EM structures of CRL2FEM1B bound to different C-degrons, we elucidate the dimeric assembly of the complex. Furthermore, we reveal distinct dimerization states of unmodified and neddylated CRL2FEM1B to uncover the NEDD8-mediated activation mechanism of CRL2FEM1B. Our research also indicates that, FEM1B utilizes a bipartite mechanism to recognize both the C-terminal proline and an upstream aromatic residue within the substrate. These structural findings, complemented by in vitro ubiquitination and in vivo cell-based assays, demonstrate that CRL2FEM1B-mediated polyubiquitination and subsequent protein turnover depend on both FEM1B-degron interactions and the dimerization state of the E3 ligase complex. Overall, this study deepens our molecular understanding of how Cullin-RING E3 ligase substrate selection mediates protein turnover.
Collapse
Affiliation(s)
- Xinyan Chen
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China
| | - Anat Raiff
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shanshan Li
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China
| | - Qiong Guo
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China
| | - Jiahai Zhang
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China
| | - Hualin Zhou
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China
| | - Richard T Timms
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China
| | - Stephen J Elledge
- Division of Genetics, Department of Medicine, Howard Hughes Medical Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Itay Koren
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel.
| | - Kaiming Zhang
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China.
| | - Chao Xu
- MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, PR China.
| |
Collapse
|
26
|
Nguyen DM, Rath DH, Devost D, Pétrin D, Rizk R, Ji AX, Narayanan N, Yong D, Zhai A, Kuntz DA, Mian MUQ, Pomroy NC, Keszei AFA, Benlekbir S, Mazhab-Jafari MT, Rubinstein JL, Hébert TE, Privé GG. Structure and dynamics of a pentameric KCTD5/CUL3/Gβγ E3 ubiquitin ligase complex. Proc Natl Acad Sci U S A 2024; 121:e2315018121. [PMID: 38625940 PMCID: PMC11047111 DOI: 10.1073/pnas.2315018121] [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: 09/21/2023] [Accepted: 03/07/2024] [Indexed: 04/18/2024] Open
Abstract
Heterotrimeric G proteins can be regulated by posttranslational modifications, including ubiquitylation. KCTD5, a pentameric substrate receptor protein consisting of an N-terminal BTB domain and a C-terminal domain, engages CUL3 to form the central scaffold of a cullin-RING E3 ligase complex (CRL3KCTD5) that ubiquitylates Gβγ and reduces Gβγ protein levels in cells. The cryo-EM structure of a 5:5:5 KCTD5/CUL3NTD/Gβ1γ2 assembly reveals a highly dynamic complex with rotations of over 60° between the KCTD5BTB/CUL3NTD and KCTD5CTD/Gβγ moieties of the structure. CRL3KCTD5 engages the E3 ligase ARIH1 to ubiquitylate Gβγ in an E3-E3 superassembly, and extension of the structure to include full-length CUL3 with RBX1 and an ARIH1~ubiquitin conjugate reveals that some conformational states position the ARIH1~ubiquitin thioester bond to within 10 Å of lysine-23 of Gβ and likely represent priming complexes. Most previously described CRL/substrate structures have consisted of monovalent complexes and have involved flexible peptide substrates. The structure of the KCTD5/CUL3NTD/Gβγ complex shows that the oligomerization of a substrate receptor can generate a polyvalent E3 ligase complex and that the internal dynamics of the substrate receptor can position a structured target for ubiquitylation in a CRL3 complex.
Collapse
Affiliation(s)
- Duc Minh Nguyen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Deanna H. Rath
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QCH3G 1Y6, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QCH3G 1Y6, Canada
| | - Robert Rizk
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QCH3G 1Y6, Canada
| | - Alan X. Ji
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Naveen Narayanan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
| | - Darren Yong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
| | - Andrew Zhai
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
| | - Douglas A. Kuntz
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
| | - Maha U. Q. Mian
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
| | - Neil C. Pomroy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
| | | | - Samir Benlekbir
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Mohammad T. Mazhab-Jafari
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5G 2M9, Canada
| | - John L. Rubinstein
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5G 2M9, Canada
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QCH3G 1Y6, Canada
| | - Gilbert G. Privé
- Princess Margaret Cancer Centre, University Health Network, Toronto, ONM5G 1L7, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5G 2M9, Canada
| |
Collapse
|
27
|
Wang T, Li X, Ma R, Sun J, Huang S, Sun Z, Wang M. Advancements in colorectal cancer research: Unveiling the cellular and molecular mechanisms of neddylation (Review). Int J Oncol 2024; 64:39. [PMID: 38391033 PMCID: PMC10919758 DOI: 10.3892/ijo.2024.5627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Neddylation, akin to ubiquitination, represents a post‑translational modification of proteins wherein neural precursor cell‑expressed developmentally downregulated protein 8 (NEDD8) is modified on the substrate protein through a series of reactions. Neddylation plays a pivotal role in the growth and proliferation of animal cells. In colorectal cancer (CRC), it predominantly contributes to the proliferation, metastasis and survival of tumor cells, decreasing overall patient survival. The strategic manipulation of the NEDD8‑mediated neddylation pathway holds immense therapeutic promise in terms of the potential to modulate the growth of tumors by regulating diverse biological responses within cancer cells, such as DNA damage response and apoptosis, among others. MLN4924 is an inhibitor of NEDD8, and its combined use with platinum drugs and irinotecan, as well as cycle inhibitors and NEDD activating enzyme inhibitors screened by drug repurposing, has been found to exert promising antitumor effects. The present review summarizes the recent progress made in the understanding of the role of NEDD8 in the advancement of CRC, suggesting that NEDD8 is a promising anti‑CRC target.
Collapse
Affiliation(s)
- Tianyu Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Xiaobing Li
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Ruijie Ma
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Jian Sun
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, P.R. China
| | - Shuhong Huang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
- Science and Technology Innovation Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Zhigang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Meng Wang
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
28
|
Jin B, Moududee SA, Ge D, Zhou P, Wang AR, Liu YZ, You Z. SCF FBXW11 Complex Targets Interleukin-17 Receptor A for Ubiquitin-Proteasome-Mediated Degradation. Biomedicines 2024; 12:755. [PMID: 38672111 PMCID: PMC11047997 DOI: 10.3390/biomedicines12040755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Interleukin-17 (IL-17) is a pro-inflammatory cytokine that participates in innate and adaptive immune responses and plays an important role in host defense, autoimmune diseases, tissue regeneration, metabolic regulation, and tumor progression. Post-translational modifications (PTMs) are crucial for protein function, stability, cellular localization, cellular transduction, and cell death. However, PTMs of IL-17 receptor A (IL-17RA) have not been investigated. Here, we show that human IL-17RA was targeted by F-box and WD repeat domain-containing 11 (FBXW11) for ubiquitination, followed by proteasome-mediated degradation. We used bioinformatics tools and biochemical techniques to determine that FBXW11 ubiquitinated IL-17RA through a lysine 27-linked polyubiquitin chain, targeting IL-17RA for proteasomal degradation. Domain 665-804 of IL-17RA was critical for interaction with FBXW11 and subsequent ubiquitination. Our study demonstrates that FBXW11 regulates IL-17 signaling pathways at the IL-17RA level.
Collapse
Affiliation(s)
- Ben Jin
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70112, USA; (B.J.); (S.A.M.)
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA 70112, USA
| | - Sayed Ala Moududee
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70112, USA; (B.J.); (S.A.M.)
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA 70112, USA
| | - Dongxia Ge
- Department of Orthopaedic Surgery, Tulane University, New Orleans, LA 70112, USA;
| | - Pengbo Zhou
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Alun R. Wang
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, Tulane University, New Orleans, LA 70112, USA;
| | - Zongbing You
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70112, USA; (B.J.); (S.A.M.)
- Department of Structural & Cellular Biology, Tulane University, New Orleans, LA 70112, USA
- Department of Orthopaedic Surgery, Tulane University, New Orleans, LA 70112, USA;
- Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University, New Orleans, LA 70112, USA
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, LA 70112, USA
- Tulane Center for Aging, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
29
|
Wang R, Huang K, Feng Y, Duan J, Ying H, Shi Q, Zhang Y, Jiang R, Yang L. Exo-miR-144-3p as a promising diagnostic biomarker for depressive symptoms in heart failure. Neurobiol Dis 2024; 192:106415. [PMID: 38266934 DOI: 10.1016/j.nbd.2024.106415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/11/2024] [Accepted: 01/21/2024] [Indexed: 01/26/2024] Open
Abstract
AIMS The prevalence of depression is higher in heart failure (HF) patients. Early screening of depressive symptoms in HF patients and timely intervention can help to improve patients' quality of life and prognosis. This study aims to explore diagnostic biomarkers by examining the expression profile of serum exosomal miRNAs in HF patients with depressive symptoms. METHODS Serum exosomal RNA was isolated and extracted from 6 HF patients with depressive symptoms (HF-DS) and 6 HF patients without depressive symptoms (HF-NDS). High-throughput sequencing was performed to obtain miRNA expression profiles and target genes were predicted for the screened differentially expressed miRNAs. Biological functions of the target genes were analyzed through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Subsequently, we collected serum exosomal RNAs from HF-DS (n = 20) and HF-NDS (n = 20). The differentially expressed miRNAs selected from the sequencing results were validated using reverse transcription quantitative polymerase chain reaction (RT-qPCR). Finally, the diagnostic efficacy of the differentially expressed exosomal miRNAs for HF-DS was evaluated by using receiver operating characteristic (ROC) curves. RESULTS A total of 19 significantly differentially expressed exosomal miRNAs were screened by high-throughput sequencing, consisting of 12 up-regulated and 7 down-regulated exosomal miRNAs. RT-qPCR validation demonstrated that the expression level of exo-miR-144-3p was significantly down-regulated in the HF-DS group, and the expression levels of exo-miR-625-3p and exo-miR-7856-5p were significantly up-regulated. In addition, the expression level of exo-miR-144-3p was negatively correlated with the severity of depressive symptoms in HF patients, and that the area under the curve (AUC) of exo-miR-144-3p for diagnosing HF-DS was 0.763. CONCLUSIONS In this study, we examined the serum exosomal miRNA expression profiles of HF patients with depressive symptoms and found that lower level of exo-miR-144-3p was associated with more severe depressive symptoms. Exo-miR-144-3p is a potential biomarker for the diagnosis of HF-DS.
Collapse
Affiliation(s)
- Ruting Wang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Kai Huang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yuehua Feng
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jiahao Duan
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Hangfeng Ying
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Qianyuan Shi
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yi Zhang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Riyue Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| |
Collapse
|
30
|
Kokic G, Yakoub G, van den Heuvel D, Wondergem AP, van der Meer PJ, van der Weegen Y, Chernev A, Fianu I, Fokkens TJ, Lorenz S, Urlaub H, Cramer P, Luijsterburg MS. Structural basis for RNA polymerase II ubiquitylation and inactivation in transcription-coupled repair. Nat Struct Mol Biol 2024; 31:536-547. [PMID: 38316879 PMCID: PMC10948364 DOI: 10.1038/s41594-023-01207-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/21/2023] [Indexed: 02/07/2024]
Abstract
During transcription-coupled DNA repair (TCR), RNA polymerase II (Pol II) transitions from a transcriptionally active state to an arrested state that allows for removal of DNA lesions. This transition requires site-specific ubiquitylation of Pol II by the CRL4CSA ubiquitin ligase, a process that is facilitated by ELOF1 in an unknown way. Using cryogenic electron microscopy, biochemical assays and cell biology approaches, we found that ELOF1 serves as an adaptor to stably position UVSSA and CRL4CSA on arrested Pol II, leading to ligase neddylation and activation of Pol II ubiquitylation. In the presence of ELOF1, a transcription factor IIS (TFIIS)-like element in UVSSA gets ordered and extends through the Pol II pore, thus preventing reactivation of Pol II by TFIIS. Our results provide the structural basis for Pol II ubiquitylation and inactivation in TCR.
Collapse
Affiliation(s)
- Goran Kokic
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Division of Structural Biology and Protein Therapeutics, Odyssey Therapeutics GmbH, Frankfurt am Main, Germany
| | - George Yakoub
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Diana van den Heuvel
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Annelotte P Wondergem
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Paula J van der Meer
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Yana van der Weegen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Aleksandar Chernev
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Isaac Fianu
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Thornton J Fokkens
- Ubiquitin Signaling Specificity, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sonja Lorenz
- Ubiquitin Signaling Specificity, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics Group, University Medical Center Göttingen, Institute of Clinical Chemistry, Göttingen, Germany
| | - Patrick Cramer
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Martijn S Luijsterburg
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
31
|
Dai Z, Liang L, Wang W, Zuo P, Yu S, Liu Y, Zhao X, Lu Y, Jin Y, Zhang F, Ding D, Deng W, Yin Y. Structural insights into the ubiquitylation strategy of the oligomeric CRL2 FEM1B E3 ubiquitin ligase. EMBO J 2024; 43:1089-1109. [PMID: 38360992 PMCID: PMC10943247 DOI: 10.1038/s44318-024-00047-y] [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: 08/22/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
Cullin-RING E3 ubiquitin ligase (CRL) family members play critical roles in numerous biological processes and diseases including cancer and Alzheimer's disease. Oligomerization of CRLs has been reported to be crucial for the regulation of their activities. However, the structural basis for its regulation and mechanism of its oligomerization are not fully known. Here, we present cryo-EM structures of oligomeric CRL2FEM1B in its unneddylated state, neddylated state in complex with BEX2 as well as neddylated state in complex with FNIP1/FLCN. These structures reveal that asymmetric dimerization of N8-CRL2FEM1B is critical for the ubiquitylation of BEX2 while FNIP1/FLCN is ubiquitylated by monomeric CRL2FEM1B. Our data present an example of the asymmetric homo-dimerization of CRL. Taken together, this study sheds light on the ubiquitylation strategy of oligomeric CRL2FEM1B according to substrates with different scales.
Collapse
Affiliation(s)
- Zonglin Dai
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ling Liang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Weize Wang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Peng Zuo
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Shang Yu
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yaqi Liu
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Xuyang Zhao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yishuo Lu
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yan Jin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Fangting Zhang
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Dian Ding
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Weiwei Deng
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
32
|
Wang X, Zhao M, Chang Y, Guan S, Li M, Yang H, Sun M. Identification of novel benzothiazole derivatives as inhibitors of NEDDylation pathway to inhibit the progression of gastric cancer. Bioorg Med Chem Lett 2024; 100:129647. [PMID: 38320715 DOI: 10.1016/j.bmcl.2024.129647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/16/2024]
Abstract
The overexpression of neddylation modification is frequently observed in human tumor cells. Targeting the neddylation pathway has been recognized as a promising anticancer therapeutic strategy, thus discovering potent and selective neddylation inhibitors is of great importance. In this study, we designed and synthesized a series of novel neddylation inhibitors bearing benzothiazole scaffolds by molecular hybridization strategy and all compounds were evaluated for antiproliferative activity against MGC-803, MCF-7, A549 and KYSE-30 cell lines. In vitro anti-tumor studies showed that the most promising compound X-10, effectively suppressed MGC-803 cells growth and migration, induced apoptosis and arrested cell cycle at G2/M phase. Importantly, by directly interacting with NAE1, X-10 blocked NAE1 activity, specifically preventing neddylation of Cullin 3 and Cullin 1, and produced a dose-response decline in the level of UBC12-NEDD8 complex. Overall, our data indicate that X-10 inhibits the process of neddylation, making it a potentially agent for both cancer prevention and therapy purposes.
Collapse
Affiliation(s)
- Xuan Wang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Mei Zhao
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuanyuan Chang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Sumeng Guan
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Mengyu Li
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hua Yang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Moran Sun
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
33
|
Hong SH, Divakaran A, Osa A, Huang OW, Wertz IE, Nomura DK. Exploiting the Cullin E3 Ligase Adaptor Protein SKP1 for Targeted Protein Degradation. ACS Chem Biol 2024; 19:442-450. [PMID: 38305738 PMCID: PMC10999000 DOI: 10.1021/acschembio.3c00642] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Targeted protein degradation with proteolysis targeting chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited substrate receptors of Cullin-RING E3 ubiquitin ligases such as cereblon and VHL. Whether core, shared, and essential components of the Cullin-RING E3 ubiquitin ligase complex can be used for PROTAC applications remains less explored. Here, we discovered a cysteine-reactive covalent recruiter EN884 against the SKP1 adapter protein of the SKP1-CUL1-F-box containing the SCF complex. We further showed that this recruiter can be used in PROTAC applications to degrade neo-substrate proteins such as BRD4 and the androgen receptor in a SKP1- and proteasome-dependent manner. Our studies demonstrate that core and essential adapter proteins within the Cullin-RING E3 ubiquitin ligase complex can be exploited for targeted protein degradation applications and that covalent chemoproteomic strategies can enable recruiter discovery against these targets.
Collapse
Affiliation(s)
- Seong Ho Hong
- Department of Chemistry, University of California, Berkeley, 2151 Berkeley Way, Room 312G, Berkeley, CA 94720 USA
- Innovative Genomics Institute, 2151 Berkeley Way, Berkeley, CA 94720 USA
| | - Anand Divakaran
- Department of Chemistry, University of California, Berkeley, 2151 Berkeley Way, Room 312G, Berkeley, CA 94720 USA
- Innovative Genomics Institute, 2151 Berkeley Way, Berkeley, CA 94720 USA
| | - Akane Osa
- Department of Chemistry, University of California, Berkeley, 2151 Berkeley Way, Room 312G, Berkeley, CA 94720 USA
- Innovative Genomics Institute, 2151 Berkeley Way, Berkeley, CA 94720 USA
| | - Oscar W. Huang
- Bristol Myers Squibb, 700 Bay Road, Redwood City, CA 94063 USA
| | - Ingrid E. Wertz
- Bristol Myers Squibb, 700 Bay Road, Redwood City, CA 94063 USA
| | - Daniel K. Nomura
- Department of Chemistry, University of California, Berkeley, 2151 Berkeley Way, Room 312G, Berkeley, CA 94720 USA
- Innovative Genomics Institute, 2151 Berkeley Way, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, 2151 Berkeley Way, Room 312G, Berkeley, Berkeley, CA 94720 USA
| |
Collapse
|
34
|
Rao S, Skulsuppaisarn M, Strong LM, Ren X, Lazarou M, Hurley JH, Hummer G. Three-step docking by WIPI2, ATG16L1, and ATG3 delivers LC3 to the phagophore. SCIENCE ADVANCES 2024; 10:eadj8027. [PMID: 38324698 PMCID: PMC10851258 DOI: 10.1126/sciadv.adj8027] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
The covalent attachment of ubiquitin-like LC3 proteins (microtubule-associated proteins 1A/1B light chain 3) prepares the autophagic membrane for cargo recruitment. We resolve key steps in LC3 lipidation by combining molecular dynamics simulations and experiments in vitro and in cellulo. We show how the E3-like ligaseautophagy-related 12 (ATG12)-ATG5-ATG16L1 in complex with the E2-like conjugase ATG3 docks LC3 onto the membrane in three steps by (i) the phosphatidylinositol 3-phosphate effector protein WD repeat domain phosphoinositide-interacting protein 2 (WIPI2), (ii) helix α2 of ATG16L1, and (iii) a membrane-interacting surface of ATG3. Phosphatidylethanolamine (PE) lipids concentrate in a region around the thioester bond between ATG3 and LC3, highlighting residues with a possible role in the catalytic transfer of LC3 to PE, including two conserved histidines. In a near-complete pathway from the initial membrane recruitment to the LC3 lipidation reaction, the three-step targeting of the ATG12-ATG5-ATG16L1 machinery establishes a high level of regulatory control.
Collapse
Affiliation(s)
- Shanlin Rao
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Marvin Skulsuppaisarn
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Lisa M. Strong
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xuefeng Ren
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael Lazarou
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - James H. Hurley
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Institute of Biophysics, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| |
Collapse
|
35
|
Barnsby-Greer L, Mabbitt PD, Dery MA, Squair DR, Wood NT, Lamoliatte F, Lange SM, Virdee S. UBE2A and UBE2B are recruited by an atypical E3 ligase module in UBR4. Nat Struct Mol Biol 2024; 31:351-363. [PMID: 38182926 PMCID: PMC10873205 DOI: 10.1038/s41594-023-01192-4] [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: 02/15/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024]
Abstract
UBR4 is a 574 kDa E3 ligase (E3) of the N-degron pathway with roles in neurodevelopment, age-associated muscular atrophy and cancer. The catalytic module that carries out ubiquitin (Ub) transfer remains unknown. Here we identify and characterize a distinct E3 module within human UBR4 consisting of a 'hemiRING' zinc finger, a helical-rich UBR zinc-finger interacting (UZI) subdomain, and an N-terminal region that can serve as an affinity factor for the E2 conjugating enzyme (E2). The structure of an E2-E3 complex provides atomic-level insight into the specificity determinants of the hemiRING toward the cognate E2s UBE2A/UBE2B. Via an allosteric mechanism, the UZI subdomain modestly activates the Ub-loaded E2 (E2∼Ub). We propose attenuated activation is complemented by the intrinsically high lysine reactivity of UBE2A, and their cooperation imparts a reactivity profile important for substrate specificity and optimal degradation kinetics. These findings reveal the mechanistic underpinnings of a neuronal N-degron E3, its specific recruitment of UBE2A, and highlight the underappreciated architectural diversity of cross-brace domains with Ub E3 activity.
Collapse
Affiliation(s)
- Lucy Barnsby-Greer
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Peter D Mabbitt
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
- Scion, Rotorua, New Zealand
| | - Marc-Andre Dery
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Daniel R Squair
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Nicola T Wood
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Frederic Lamoliatte
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Sven M Lange
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Satpal Virdee
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK.
| |
Collapse
|
36
|
Hu Y, Zhang Z, Mao Q, Zhang X, Hao A, Xun Y, Wang Y, Han L, Zhan W, Liu Q, Yin Y, Peng C, Moresco EMY, Chen Z, Beutler B, Sun L. Dynamic molecular architecture and substrate recruitment of cullin3-RING E3 ligase CRL3 KBTBD2. Nat Struct Mol Biol 2024; 31:336-350. [PMID: 38332366 PMCID: PMC11791872 DOI: 10.1038/s41594-023-01182-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/16/2023] [Indexed: 02/10/2024]
Abstract
Phosphatidylinositol 3-kinase α, a heterodimer of catalytic p110α and one of five regulatory subunits, mediates insulin- and insulin like growth factor-signaling and, frequently, oncogenesis. Cellular levels of the regulatory p85α subunit are tightly controlled by regulated proteasomal degradation. In adipose tissue and growth plates, failure of K48-linked p85α ubiquitination causes diabetes, lipodystrophy and dwarfism in mice, as in humans with SHORT syndrome. Here we elucidated the structures of the key ubiquitin ligase complexes regulating p85α availability. Specificity is provided by the substrate receptor KBTBD2, which recruits p85α to the cullin3-RING E3 ubiquitin ligase (CRL3). CRL3KBTBD2 forms multimers, which disassemble into dimers upon substrate binding (CRL3KBTBD2-p85α) and/or neddylation by the activator NEDD8 (CRL3KBTBD2~N8), leading to p85α ubiquitination and degradation. Deactivation involves dissociation of NEDD8 mediated by the COP9 signalosome and displacement of KBTBD2 by the inhibitor CAND1. The hereby identified structural basis of p85α regulation opens the way to better understanding disturbances of glucose regulation, growth and cancer.
Collapse
Affiliation(s)
- Yuxia Hu
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhao Zhang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qiyu Mao
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiang Zhang
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Aihua Hao
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yu Xun
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yeda Wang
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lin Han
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wuqiang Zhan
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qianying Liu
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Eva Marie Y Moresco
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhenguo Chen
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Lei Sun
- Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Key Laboratory of Medical Epigenetics and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Wang K, Diaz S, Li L, Lohman JR, Liu X. CAND1 inhibits Cullin-2-RING ubiquitin ligases for enhanced substrate specificity. Nat Struct Mol Biol 2024; 31:323-335. [PMID: 38177676 PMCID: PMC10923007 DOI: 10.1038/s41594-023-01167-5] [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: 03/13/2023] [Accepted: 10/29/2023] [Indexed: 01/06/2024]
Abstract
Through targeting essential cellular regulators for ubiquitination and serving as a major platform for discovering proteolysis-targeting chimera (PROTAC) drugs, Cullin-2 (CUL2)-RING ubiquitin ligases (CRL2s) comprise an important family of CRLs. The founding members of CRLs, the CUL1-based CRL1s, are known to be activated by CAND1, which exchanges the variable substrate receptors associated with the common CUL1 core and promotes the dynamic assembly of CRL1s. Here we find that CAND1 inhibits CRL2-mediated protein degradation in human cells. This effect arises due to altered binding kinetics, involving CAND1 and CRL2VHL, as we illustrate that CAND1 dramatically increases the dissociation rate of CRL2s but barely accelerates the assembly of stable CRL2s. Using PROTACs that differently recruit neo-substrates to CRL2VHL, we demonstrate that the inhibitory effect of CAND1 helps distinguish target proteins with different affinities for CRL2s, presenting a mechanism for selective protein degradation with proper pacing in the changing cellular environment.
Collapse
Affiliation(s)
- Kankan Wang
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Stephanie Diaz
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
- Department of Neurosurgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Lihong Li
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
- Center for Plant Biology, Purdue University, West Lafayette, IN, USA
| | - Jeremy R Lohman
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Xing Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA.
- Center for Plant Biology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
38
|
Chrustowicz J, Sherpa D, Li J, Langlois CR, Papadopoulou EC, Vu DT, Hehl LA, Karayel Ö, Beier V, von Gronau S, Müller J, Prabu JR, Mann M, Kleiger G, Alpi AF, Schulman BA. Multisite phosphorylation dictates selective E2-E3 pairing as revealed by Ubc8/UBE2H-GID/CTLH assemblies. Mol Cell 2024; 84:293-308.e14. [PMID: 38113892 PMCID: PMC10843684 DOI: 10.1016/j.molcel.2023.11.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/29/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023]
Abstract
Ubiquitylation is catalyzed by coordinated actions of E3 and E2 enzymes. Molecular principles governing many important E3-E2 partnerships remain unknown, including those for RING-family GID/CTLH E3 ubiquitin ligases and their dedicated E2, Ubc8/UBE2H (yeast/human nomenclature). GID/CTLH-Ubc8/UBE2H-mediated ubiquitylation regulates biological processes ranging from yeast metabolic signaling to human development. Here, cryoelectron microscopy (cryo-EM), biochemistry, and cell biology reveal this exquisitely specific E3-E2 pairing through an unconventional catalytic assembly and auxiliary interactions 70-100 Å away, mediated by E2 multisite phosphorylation. Rather than dynamic polyelectrostatic interactions reported for other ubiquitylation complexes, multiple Ubc8/UBE2H phosphorylation sites within acidic CK2-targeted sequences specifically anchor the E2 C termini to E3 basic patches. Positions of phospho-dependent interactions relative to the catalytic domains correlate across evolution. Overall, our data show that phosphorylation-dependent multivalency establishes a specific E3-E2 partnership, is antagonistic with dephosphorylation, rigidifies the catalytic centers within a flexing GID E3-substrate assembly, and facilitates substrate collision with ubiquitylation active sites.
Collapse
Affiliation(s)
- Jakub Chrustowicz
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Dawafuti Sherpa
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Jerry Li
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, NV 89154, USA
| | - Christine R Langlois
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Eleftheria C Papadopoulou
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany; Technical University of Munich, School of Natural Sciences, Munich 85748, Germany
| | - D Tung Vu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Laura A Hehl
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany; Technical University of Munich, School of Natural Sciences, Munich 85748, Germany
| | - Özge Karayel
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Viola Beier
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Susanne von Gronau
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Judith Müller
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - J Rajan Prabu
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Gary Kleiger
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, NV 89154, USA
| | - Arno F Alpi
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany; Technical University of Munich, School of Natural Sciences, Munich 85748, Germany.
| |
Collapse
|
39
|
Hong SH, Osa A, Huang OW, Wertz IE, Nomura DK. Exploiting the Cullin E3 Ligase Adaptor Protein SKP1 for Targeted Protein Degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563371. [PMID: 37904950 PMCID: PMC10614948 DOI: 10.1101/2023.10.20.563371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Targeted protein degradation with Proteolysis Targeting Chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited substrate receptors of Cullin-RING E3 ubiquitin ligases such as cereblon and VHL. Whether core, shared, and essential components of the Cullin-RING E3 ubiquitin ligase complex can be used for PROTAC applications remains less explored. Here, we discovered a cysteine-reactive covalent recruiter EN884 against the SKP1 adapter protein of the SKP1-CUL1-F-box containing SCF complex. We further showed that this recruiter can be used in PROTAC applications to degrade neo-substrate proteins such as BRD4 and the androgen receptor in a SKP1- and proteasome-dependent manner. Our studies demonstrate that core and essential adapter proteins within the Cullin-RING E3 ubiquitin ligase complex can be exploited for targeted protein degradation applications and that covalent chemoproteomic strategies can enable recruiter discovery against these targets.
Collapse
Affiliation(s)
- Seong Ho Hong
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
| | - Akane Osa
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
| | | | | | - Daniel K. Nomura
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
| |
Collapse
|
40
|
Bodrug T, Welsh KA, Bolhuis DL, Paulаkonis E, Martinez-Chacin RC, Liu B, Pinkin N, Bonacci T, Cui L, Xu P, Roscow O, Amann SJ, Grishkovskaya I, Emanuele MJ, Harrison JS, Steimel JP, Hahn KM, Zhang W, Zhong ED, Haselbach D, Brown NG. Time-resolved cryo-EM (TR-EM) analysis of substrate polyubiquitination by the RING E3 anaphase-promoting complex/cyclosome (APC/C). Nat Struct Mol Biol 2023; 30:1663-1674. [PMID: 37735619 PMCID: PMC10643132 DOI: 10.1038/s41594-023-01105-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/21/2023] [Indexed: 09/23/2023]
Abstract
Substrate polyubiquitination drives a myriad of cellular processes, including the cell cycle, apoptosis and immune responses. Polyubiquitination is highly dynamic, and obtaining mechanistic insight has thus far required artificially trapped structures to stabilize specific steps along the enzymatic process. So far, how any ubiquitin ligase builds a proteasomal degradation signal, which is canonically regarded as four or more ubiquitins, remains unclear. Here we present time-resolved cryogenic electron microscopy studies of the 1.2 MDa E3 ubiquitin ligase, known as the anaphase-promoting complex/cyclosome (APC/C), and its E2 co-enzymes (UBE2C/UBCH10 and UBE2S) during substrate polyubiquitination. Using cryoDRGN (Deep Reconstructing Generative Networks), a neural network-based approach, we reconstruct the conformational changes undergone by the human APC/C during polyubiquitination, directly visualize an active E3-E2 pair modifying its substrate, and identify unexpected interactions between multiple ubiquitins with parts of the APC/C machinery, including its coactivator CDH1. Together, we demonstrate how modification of substrates with nascent ubiquitin chains helps to potentiate processive substrate polyubiquitination, allowing us to model how a ubiquitin ligase builds a proteasomal degradation signal.
Collapse
Affiliation(s)
- Tatyana Bodrug
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kaeli A Welsh
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Derek L Bolhuis
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ethan Paulаkonis
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Raquel C Martinez-Chacin
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Bei Liu
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, China
| | - Nicholas Pinkin
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Thomas Bonacci
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Liying Cui
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Pengning Xu
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Olivia Roscow
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - Sascha Josef Amann
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Irina Grishkovskaya
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Michael J Emanuele
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Joseph S Harrison
- Department of Chemistry, University of the Pacific, Stockton, CA, USA
| | - Joshua P Steimel
- School of Engineering, California Polytechnic State University Humboldt, Arcata, CA, USA
| | - Klaus M Hahn
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Wei Zhang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - Ellen D Zhong
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - David Haselbach
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria.
| | - Nicholas G Brown
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
41
|
Tang L, Wei D, Xu X, Mo D, Cheng D, Yan F. FOXO1-regulated lncRNA CYP1B1-AS1 suppresses breast cancer cell proliferation by inhibiting neddylation. Breast Cancer Res Treat 2023; 202:397-408. [PMID: 37640964 PMCID: PMC10505597 DOI: 10.1007/s10549-023-07090-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE Overactivated neddylation is considered to be a common event in cancer. Long non-coding RNAs (lncRNAs) can regulate cancer development by mediating post-translational modifications. However, the role of lncRNA in neddylation modification remains unclear. METHODS LncRNA cytochrome P450 family 1 subfamily B member 1 antisense RNA 1 (CYP1B1-AS1) expression in breast cancer tissues was evaluated by RT-PCR and TCGA BRCA data. Gain and loss of function experiments were performed to explore the role of CYP1B1-AS1 in breast cancer cell proliferation and apoptosis in vitro and in vivo. Luciferase assay, CHIP-qPCR assay, transcriptome sequencing, RNA-pulldown assay, mass spectrometry, RIP-PCR and Western blot were used to investigate the regulatory factors of CYP1B1-AS1 expression and the molecular mechanism of CYP1B1-AS1 involved in neddylation modification. RESULTS We found that CYP1B1-AS1 was down-regulated in breast cancer tissues and correlated with prognosis. In vivo and in vitro functional experiments confirmed that CYP1B1-AS1 inhibited cell proliferation and induced apoptosis. Mechanistically, CYP1B1-AS1 was regulated by the transcription factor, forkhead box O1 (FOXO1), and could be upregulated by inhibiting the PI3K/FOXO1 pathway. Moreover, CYP1B1-AS1 bound directly to NEDD8 activating enzyme E1 subunit 1 (NAE1) to regulate protein neddylation. CONCLUSION This study reports for the first time that CYP1B1-AS1 inhibits protein neddylation to affect breast cancer cell proliferation, which provides a new strategy for the treatment of breast cancer by lncRNA targeting neddylation modification.
Collapse
Affiliation(s)
- Li Tang
- Department of Clinical Laboratory, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, No. 42 Baiziting Road, Nanjing, 210009, People's Republic of China.
| | - Da Wei
- Department of Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, People's Republic of China
| | - Xinyu Xu
- Department of Pathology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, People's Republic of China
| | - Dongping Mo
- Department of Clinical Laboratory, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, No. 42 Baiziting Road, Nanjing, 210009, People's Republic of China
| | - Daofu Cheng
- Department of Clinical Laboratory, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, No. 42 Baiziting Road, Nanjing, 210009, People's Republic of China
| | - Feng Yan
- Department of Clinical Laboratory, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, No. 42 Baiziting Road, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
42
|
Füzesi-Levi MG, Ben-Nissan G, Listov D, Fridmann Sirkis Y, Hayouka Z, Fleishman S, Sharon M. The C-terminal tail of CSNAP attenuates the CSN complex. Life Sci Alliance 2023; 6:e202201634. [PMID: 37460146 PMCID: PMC10355216 DOI: 10.26508/lsa.202201634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Protein degradation is one of the essential mechanisms that enables reshaping of the proteome landscape in response to various stimuli. The largest E3 ubiquitin ligase family that targets proteins to degradation by catalyzing ubiquitination is the cullin-RING ligases (CRLs). Many of the proteins that are regulated by CRLs are central to tumorigenesis and tumor progression, and dysregulation of the CRL family is frequently associated with cancer. The CRL family comprises ∼300 complexes, all of which are regulated by the COP9 signalosome complex (CSN). Therefore, CSN is considered an attractive target for therapeutic intervention. Research efforts for targeted CSN inhibition have been directed towards inhibition of the complex enzymatic subunit, CSN5. Here, we have taken a fresh approach focusing on CSNAP, the smallest CSN subunit. Our results show that the C-terminal region of CSNAP is tightly packed within the CSN complex, in a groove formed by CSN3 and CSN8. We show that a 16 amino acid C-terminal peptide, derived from this CSN-interacting region, can displace the endogenous CSNAP subunit from the complex. This, in turn, leads to a CSNAP null phenotype that attenuates CSN activity and consequently CRLs function. Overall, our findings emphasize the potential of a CSNAP-based peptide for CSN inhibition as a new therapeutic avenue.
Collapse
Affiliation(s)
- Maria G Füzesi-Levi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Gili Ben-Nissan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Dina Listov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Sarel Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
43
|
Choudhary D, Kaur A, Singh P, Chaudhary G, Kaur R, Bayan MF, Chandrasekaran B, Marji SM, Ayman R. Target protein degradation by protacs: A budding cancer treatment strategy. Pharmacol Ther 2023; 250:108525. [PMID: 37696366 DOI: 10.1016/j.pharmthera.2023.108525] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023]
Abstract
Cancer is one of the most common causes of death. So, its lethal effect increases with time. Near about hundreds of cancers are known in humans. Cancer treatment is done to cure or prolonged remission, and shrinkage of the tumor. Cytotoxic agents, biological agents/targeted drugs, hormonal drugs, surgery, radiotherapy/proton therapy, chemotherapy, immunotherapy, and gene therapy are currently used in the treatment of cancer but their cost is high and cause various side effects. Seeing this, some new targeted strategies such as PROTACs are the need of the time. Proteolysis targeting chimera (PROTAC) has become one of the most discussed topics regarding cancer treatment. Few of the PROTAC molecules are in the trial phases. PROTACs have many advantages over other strategies such as modularity, compatibility, sub-stoichiometric activity, acting on undruggable targets, molecular design, and acts on intracellular targets, selectivity and specificity can be recruited for any cancer, versatility, and others. PROTACs are having some unclear questions on their pharmacokinetics, heavy-molecular weight, etc. PROTACs are anticipated to bring about a conversion in current healthcare and will emerge as booming treatments. In this review article we summarize PROTACs, their mechanism of action, uses, advantages, disadvantages, challenges, and future aspects for the successful development of potent PROTACs as a drug strategy.
Collapse
Affiliation(s)
- Diksha Choudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Amritpal Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Pargat Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Gaurav Chaudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | - Mohammad F Bayan
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | | | - Saeed M Marji
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | - Reema Ayman
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| |
Collapse
|
44
|
Roberts CG, Franklin TG, Pruneda JN. Ubiquitin-targeted bacterial effectors: rule breakers of the ubiquitin system. EMBO J 2023; 42:e114318. [PMID: 37555693 PMCID: PMC10505922 DOI: 10.15252/embj.2023114318] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023] Open
Abstract
Regulation through post-translational ubiquitin signaling underlies a large portion of eukaryotic biology. This has not gone unnoticed by invading pathogens, many of which have evolved mechanisms to manipulate or subvert the host ubiquitin system. Bacteria are particularly adept at this and rely heavily upon ubiquitin-targeted virulence factors for invasion and replication. Despite lacking a conventional ubiquitin system of their own, many bacterial ubiquitin regulators loosely follow the structural and mechanistic rules established by eukaryotic ubiquitin machinery. Others completely break these rules and have evolved novel structural folds, exhibit distinct mechanisms of regulation, or catalyze foreign ubiquitin modifications. Studying these interactions can not only reveal important aspects of bacterial pathogenesis but also shed light on unexplored areas of ubiquitin signaling and regulation. In this review, we discuss the methods by which bacteria manipulate host ubiquitin and highlight aspects that follow or break the rules of ubiquitination.
Collapse
Affiliation(s)
- Cameron G Roberts
- Department of Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Tyler G Franklin
- Department of Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Jonathan N Pruneda
- Department of Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| |
Collapse
|
45
|
Asmar AJ, Abrams SR, Hsin J, Collins JC, Yazejian RM, Wu Y, Cho J, Doyle AD, Cinthala S, Simon M, van Jaarsveld RH, Beck DB, Kerosuo L, Werner A. A ubiquitin-based effector-to-inhibitor switch coordinates early brain, craniofacial, and skin development. Nat Commun 2023; 14:4499. [PMID: 37495603 PMCID: PMC10371987 DOI: 10.1038/s41467-023-40223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 07/18/2023] [Indexed: 07/28/2023] Open
Abstract
The molecular mechanisms that coordinate patterning of the embryonic ectoderm into spatially distinct lineages to form the nervous system, epidermis, and neural crest-derived craniofacial structures are unclear. Here, biochemical disease-variant profiling reveals a posttranslational pathway that drives early ectodermal differentiation in the vertebrate head. The anteriorly expressed ubiquitin ligase CRL3-KLHL4 restricts signaling of the ubiquitous cytoskeletal regulator CDC42. This regulation relies on the CDC42-activating complex GIT1-βPIX, which CRL3-KLHL4 exploits as a substrate-specific co-adaptor to recognize and monoubiquitylate PAK1. Surprisingly, we find that ubiquitylation converts the canonical CDC42 effector PAK1 into a CDC42 inhibitor. Loss of CRL3-KLHL4 or a disease-associated KLHL4 variant reduce PAK1 ubiquitylation causing overactivation of CDC42 signaling and defective ectodermal patterning and neurulation. Thus, tissue-specific restriction of CDC42 signaling by a ubiquitin-based effector-to-inhibitor is essential for early face, brain, and skin formation, revealing how cell-fate and morphometric changes are coordinated to ensure faithful organ development.
Collapse
Affiliation(s)
- Anthony J Asmar
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shaun R Abrams
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jenny Hsin
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jason C Collins
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rita M Yazejian
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Youmei Wu
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jean Cho
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew D Doyle
- NIDCR Imaging Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Samhitha Cinthala
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marleen Simon
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - David B Beck
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY, USA
| | - Laura Kerosuo
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Achim Werner
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
46
|
Ying S, Guo Q, Zhang C. KLHL3-dependent WNK4 degradation affected by potassium through the neddylation and autophagy pathway. BMC Nephrol 2023; 24:217. [PMID: 37481568 PMCID: PMC10362690 DOI: 10.1186/s12882-023-03257-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/29/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Studies reported that kelch-like protein 3 (KLHL3)-Cullin3(CUL3) E3 ligase ubiquitinated with-no-lysine kinase 4 (WNK4). Impaired WNK4 ubiquitination plays a key role in Familial hyperkalemic hypertension (FHHt, also called pseudohypoaldosteronism type II) which results from overaction of thiazide-sensitive sodium chloride cotransport (NCC). In addition, researchers have also found that dietary potassium deficiency activates NCC along the renal distal convoluted tubule (DCT). However, the underlying mechanism remains unclear about the relationship between potassium and WNK4. METHODS In the present study, we conducted in vitro and in vivo experiments to confirm that KLHL3-dependent WNK4 degradation is affected by potassium through the neddylation and autophagy pathway. In vitro, the WNK4 and KLHL3 plasmids were cotransfected into HEK293 cell lines by lipofectamine 2000, and then incubated with different potassium concentrations (1mmol/L and 10mmol/L) for 24 h, and further treated with MLN4924 or the autophagy inhibitor or both of MLN4924 and the autophagy inhibitor for another 24 h respectively. In vivo, we created mice that were fed with low or high potassium diets and then were injected MLN4924 in the experimental groups. The expression of WNK4, pWNK4, KLHL3, NEDD8, LC3 ,and P62 was detected by western blotting in vitro and vivo experiments. RESULTS We found that the abundance and phosphorylation of WNK4 increase when neddylation is inhibited both in vitro and vivo. Furthermore, the abundance of pWNK4, WNK4, NEDD8, and KLHL3 was increased in the low potassium (LK) group. Inhibiting autophagy can ameliorate the effect of potassium on the abundance and activity of WNK4 to some extent. CONCLUSION These findings suggest a complex regulation of potassium in the degradation of WNK4. Low potassium can activate WNK4, which may be related to neddylation and autophagy, but the mechanism needs to be further studied.
Collapse
Affiliation(s)
- Siqi Ying
- Department of Nephrology, Jing'an District Center Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Qin Guo
- Department of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Shi, China
| | - Chong Zhang
- Department of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Shi, China.
| |
Collapse
|
47
|
Gu J, Ke P, Guo H, Liu J, Liu Y, Tian X, Huang Z, Xu X, Xu D, Ma Y, Wang X, Xiao F. KCTD13-mediated ubiquitination and degradation of GluN1 regulates excitatory synaptic transmission and seizure susceptibility. Cell Death Differ 2023; 30:1726-1741. [PMID: 37142655 PMCID: PMC10307852 DOI: 10.1038/s41418-023-01174-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common and severe form of epilepsy in adults; however, its underlying pathomechanisms remain elusive. Dysregulation of ubiquitination is increasingly recognized to contribute to the development and maintenance of epilepsy. Herein, we observed for the first time that potassium channel tetramerization domain containing 13 (KCTD13) protein, a substrate-specific adapter for cullin3-based E3 ubiquitin ligase, was markedly down-regulated in the brain tissue of patients with TLE. In a TLE mouse model, the protein expression of KCTD13 dynamically changed during epileptogenesis. Knockdown of KCTD13 in the mouse hippocampus significantly enhanced seizure susceptibility and severity, whereas overexpression of KCTD13 showed the opposite effect. Mechanistically, GluN1, an obligatory subunit of N-methyl-D-aspartic acid receptors (NMDARs), was identified as a potential substrate protein of KCTD13. Further investigation revealed that KCTD13 facilitates lysine-48-linked polyubiquitination of GluN1 and its degradation through the ubiquitin-proteasome pathway. Besides, the lysine residue 860 of GluN1 is the main ubiquitin site. Importantly, dysregulation of KCTD13 affected membrane expression of glutamate receptors and impaired glutamate synaptic transmission. Systemic administration of the NMDAR inhibitor memantine significantly rescued the epileptic phenotype aggravated by KCTD13 knockdown. In conclusion, our results demonstrated an unrecognized pathway of KCTD13-GluN1 in epilepsy, suggesting KCTD13 as a potential neuroprotective therapeutic target for epilepsy.
Collapse
Affiliation(s)
- Juan Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Pingyang Ke
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Haokun Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Jing Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Yan Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Xin Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Demei Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Yuanlin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
| | - Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
- Institute for Brain Science and Disease of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
48
|
Chauhan C, Kaundal RK. Understanding the role of cGAS-STING signaling in ischemic stroke: a new avenue for drug discovery. Expert Opin Drug Discov 2023; 18:1133-1149. [PMID: 37537969 DOI: 10.1080/17460441.2023.2244409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
INTRODUCTION Ischemic stroke is a significant global health challenge with limited treatment options. Neuroinflammation, driven by microglial activation, plays a critical role in stroke pathophysiology. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has emerged as a key player in microglial activation, sterile neuroinflammation, and cell death following stroke. Understanding the interplay between this pathway and stroke pathophysiology is crucial for exploring newer therapeutics for stroke patients. AREAS COVERED This review discusses the pivotal role of the cGAS-STING pathway in ischemic stroke. It explores the interplay between cGAS-STING activation, neuroinflammation, microglia activation, M2 polarization, neutrophil infiltration, and cytokine release. Additionally, the authors examine its contributions to various cell death programs (pyroptosis, apoptosis, necroptosis, lysosomal cell death, autophagy, and ferroptosis). The review summarizes recent studies on targeting cGAS-STING signaling in stroke, highlighting the therapeutic potential of small molecule inhibitors and RNA-based approaches in mitigating neuroinflammation, preventing cell death, and improving patient outcomes. EXPERT OPINION Understanding cGAS-STING signaling in ischemic stroke offers an exciting avenue for drug discovery. Targeting this pathway holds promise for developing novel therapeutics that effectively mitigate neuroinflammation, prevent cell death, and enhance patient outcomes. Further research and development of therapeutic strategies are warranted to fully exploit the potential of this pathway as a therapeutic target for stroke.
Collapse
Affiliation(s)
- Chandan Chauhan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, India
| | - Ravinder Kumar Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, India
| |
Collapse
|
49
|
Su Y, Luo Y, Zhang P, Lin H, Pu W, Zhang H, Wang H, Hao Y, Xiao Y, Zhang X, Wei X, Nie S, Zhang K, Fu Q, Chen H, Huang N, Ren Y, Wu M, Chow BKC, Chen X, Jin W, Wang F, Zhao L, Rao F. Glucose-induced CRL4 COP1-p53 axis amplifies glycometabolism to drive tumorigenesis. Mol Cell 2023:S1097-2765(23)00432-X. [PMID: 37390815 DOI: 10.1016/j.molcel.2023.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 04/19/2023] [Accepted: 06/06/2023] [Indexed: 07/02/2023]
Abstract
The diabetes-cancer association remains underexplained. Here, we describe a glucose-signaling axis that reinforces glucose uptake and glycolysis to consolidate the Warburg effect and overcome tumor suppression. Specifically, glucose-dependent CK2 O-GlcNAcylation impedes its phosphorylation of CSN2, a modification required for the deneddylase CSN to sequester Cullin RING ligase 4 (CRL4). Glucose, therefore, elicits CSN-CRL4 dissociation to assemble the CRL4COP1 E3 ligase, which targets p53 to derepress glycolytic enzymes. A genetic or pharmacologic disruption of the O-GlcNAc-CK2-CSN2-CRL4COP1 axis abrogates glucose-induced p53 degradation and cancer cell proliferation. Diet-induced overnutrition upregulates the CRL4COP1-p53 axis to promote PyMT-induced mammary tumorigenesis in wild type but not in mammary-gland-specific p53 knockout mice. These effects of overnutrition are reversed by P28, an investigational peptide inhibitor of COP1-p53 interaction. Thus, glycometabolism self-amplifies via a glucose-induced post-translational modification cascade culminating in CRL4COP1-mediated p53 degradation. Such mutation-independent p53 checkpoint bypass may represent the carcinogenic origin and targetable vulnerability of hyperglycemia-driven cancer.
Collapse
Affiliation(s)
- Yang Su
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yifan Luo
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China; School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Peitao Zhang
- Department of Thyroid and Neck Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Hong Lin
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Weijie Pu
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hongyun Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Huifang Wang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yi Hao
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yihang Xiao
- School of Science, Westlake University, Westlake Laboratory of Life Sciences and Biomedicine, and Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiaozhe Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiayun Wei
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Siyue Nie
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Keren Zhang
- BGI-Shenzhen, Beishan Industrial Zone 11th building, Yantian District, Shenzhen, Guangdong, 518083, China
| | - Qiuyu Fu
- National Institute of Biological Sciences, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Hao Chen
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Niu Huang
- National Institute of Biological Sciences, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Yan Ren
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingxuan Wu
- School of Science, Westlake University, Westlake Laboratory of Life Sciences and Biomedicine, and Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | | | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Wenfei Jin
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China.
| | - Li Zhao
- Department of Thyroid and Neck Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| | - Feng Rao
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
50
|
Blackburn PR, Ebstein F, Hsieh TC, Motta M, Radio FC, Herkert JC, Rinne T, Thiffault I, Rapp M, Alders M, Maas S, Gerard B, Smol T, Vincent-Delorme C, Cogné B, Isidor B, Vincent M, Bachmann-Gagescu R, Rauch A, Joset P, Ferrero GB, Ciolfi A, Husson T, Guerrot AM, Bacino C, Macmurdo C, Thompson SS, Rosenfeld JA, Faivre L, Mau-Them FT, Deb W, Vignard V, Agrawal PB, Madden JA, Goldenberg A, Lecoquierre F, Zech M, Prokisch H, Necpál J, Jech R, Winkelmann J, Koprušáková MT, Konstantopoulou V, Younce JR, Shinawi M, Mighton C, Fung C, Morel C, Ellis JL, DiTroia S, Barth M, Bonneau D, Krapels I, Stegmann S, van der Schoot V, Brunet T, Bußmann C, Mignot C, Courtin T, Ravelli C, Keren B, Ziegler A, Hasadsri L, Pichurin PN, Klee EW, Grand K, Sanchez-Lara PA, Krüger E, Bézieau S, Klinkhammer H, Krawitz PM, Eichler EE, Tartaglia M, Küry S, Wang T. Loss-of-function variants in CUL3 cause a syndromic neurodevelopmental disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.13.23290941. [PMID: 37398376 PMCID: PMC10312857 DOI: 10.1101/2023.06.13.23290941] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Purpose De novo variants in CUL3 (Cullin-3 ubiquitin ligase) have been strongly associated with neurodevelopmental disorders (NDDs), but no large case series have been reported so far. Here we aimed to collect sporadic cases carrying rare variants in CUL3, describe the genotype-phenotype correlation, and investigate the underlying pathogenic mechanism. Methods Genetic data and detailed clinical records were collected via multi-center collaboration. Dysmorphic facial features were analyzed using GestaltMatcher. Variant effects on CUL3 protein stability were assessed using patient-derived T-cells. Results We assembled a cohort of 35 individuals with heterozygous CUL3 variants presenting a syndromic NDD characterized by intellectual disability with or without autistic features. Of these, 33 have loss-of-function (LoF) and two have missense variants. CUL3 LoF variants in patients may affect protein stability leading to perturbations in protein homeostasis, as evidenced by decreased ubiquitin-protein conjugates in vitro . Specifically, we show that cyclin E1 (CCNE1) and 4E-BP1 (EIF4EBP1), two prominent substrates of CUL3, fail to be targeted for proteasomal degradation in patient-derived cells. Conclusion Our study further refines the clinical and mutational spectrum of CUL3 -associated NDDs, expands the spectrum of cullin RING E3 ligase-associated neuropsychiatric disorders, and suggests haploinsufficiency via LoF variants is the predominant pathogenic mechanism.
Collapse
|