1
|
Okatsu K, Kawaguchi T, Watanabe K, Taguchi Y, Takeuchi R, Okamoto A, Iwasa Y, Tomita T, Saeki Y, Sato Y, Narumi T, Fukai S. Adaptor-Specific Peptide Inhibitors of the Ubiquitin-Chain-Dependent Unfolding Activity of the Human p97(VCP)-UFD1-NPL4 Complex. J Med Chem 2025. [PMID: 40421687 DOI: 10.1021/acs.jmedchem.5c00201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
The AAA-ATPase p97, a key component of the ubiquitin-proteasome system (UPS), collaborates with its cofactor, the UFD1-NPL4 (UN) heterodimer, to unfold ubiquitinated substrates leading to proteasomal degradation. In this study, we report the development of novel peptide inhibitors that specifically target the p97-UN complex. These inhibitors are designed based on the NPL4-binding motif (NBM) of UFD1 and disrupt the interaction between p97 and the UN heterodimer. Our results demonstrate that these peptides effectively inhibit the unfolding activity of p97-UN, suggesting their potential as a therapeutic strategy for diseases associated with UPS dysfunction, such as cancer and neurodegenerative disorders. This work provides the first mechanistic insights into the inhibition of p97-UN by high-affinity peptide inhibitors and introduces promising candidates for drug development targeting the stable p97-UN complex in cells.
Collapse
Affiliation(s)
- Kei Okatsu
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Takaya Kawaguchi
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Kohei Watanabe
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Yoshinori Taguchi
- Graduate School of Medical Photonics, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Reon Takeuchi
- Graduate School of Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Akinori Okamoto
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Yasuyuki Iwasa
- Division of Protein Metabolism, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Takuya Tomita
- Division of Protein Metabolism, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yasushi Saeki
- Division of Protein Metabolism, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yusuke Sato
- Center for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Tetsuo Narumi
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
- Graduate School of Medical Photonics, Shizuoka University, Hamamatsu 432-8561, Japan
- Graduate School of Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Shuya Fukai
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
2
|
McFarland MR, Kulathu Y. Emerging tools and methods to study cell signalling mediated by branched ubiquitin chains. Biochem Soc Trans 2025:BST20253015. [PMID: 40380883 DOI: 10.1042/bst20253015] [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: 01/21/2025] [Accepted: 04/30/2025] [Indexed: 05/19/2025]
Abstract
Branched ubiquitin chains are complex molecular structures in which two or more ubiquitin moieties are attached to distinct lysine residues of a single ubiquitin molecule within a polyubiquitin chain. These bifurcated architectures significantly expand the signalling capacity of the ubiquitin system. Although branched chains constitute a substantial fraction of cellular polyubiquitin, their biological functions largely remain enigmatic due to their complex nature and the associated technical challenges of studying them. Recent technological innovations have enabled the identification of key molecular players and revealed essential roles for branched chains in diverse cellular processes. In this review, we discuss the bespoke strategies that have driven these discoveries, as well as the technologies needed to advance this rapidly evolving field.
Collapse
Affiliation(s)
- Matthew R McFarland
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Scotland, U.K
| | - Yogesh Kulathu
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Scotland, U.K
| |
Collapse
|
3
|
Skóra B, Szychowski KA. Proteostasis and autophagy disruption by the aging-related VGVAPG hexapeptide - preliminary insights into a potential novel elastin-induced neurodegeneration pathway in an in vitro human cellular neuron model. Neurochem Int 2025; 187:105992. [PMID: 40348194 DOI: 10.1016/j.neuint.2025.105992] [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: 02/17/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
The hexapeptide Val-Gly-Val-Ala-Pro-Gly (VGVAPG) is the most readily released product of elastin degradation, a process closely associated with aging. Recent studies have demonstrated the ability of this peptide to upregulate Sirtuin 2 (SIRT2) mRNA and protein expression. The correlation between HRD1 ligase (Synoviolin 1) and the degradation of SIRT2 has been previously reported in the literature. This study aimed to explore the impact of VGVAPG-induced interaction between HRD1 and SIRT2 and its effects on autophagy in differentiated SH-SY5Y cells in vitro (a simplified model of neurons). The results revealed that VGVAPG decreases HRD1 mRNA and protein expression while correlating with SIRT2 overexpression. Further analysis showed reduced SEL1L protein levels and an increase in p97/VCP protein expression. Additionally, enhanced phosphorylation of IRE1α indicated induction of ER stress in the tested cell model without affecting mTOR. Decreased proteasome activity and accumulation of ubiquitin were also noted. This phenomenon triggered VGVAPG-induced autophagy, as evidenced by increased expression of autophagy-related proteins ATG16L1, ATG5, ATG18, and FIP200. However, autophagy was suppressed probably as a result of VGVAPG-induced phosphorylation of ERK1/2. These findings demonstrate that the aging-related hexapeptide VGVAPG downregulates the function of the SEL1L-HRD1 complex, leading to SIRT2 accumulation and subsequent ER stress due to ERAD and UPS. This cascade, in turn, activates autophagy as an alternative clearance pathway aimed at restoring proteostasis; however, the process becomes dysregulated, leading to persistent ER stress. This dual effect may have significant implications in neurobiology, given the well-established correlation between autophagy impairment and aging-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszów, St. Sucharskiego 2, 35 -225, Rzeszów, Poland.
| | - Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszów, St. Sucharskiego 2, 35 -225, Rzeszów, Poland
| |
Collapse
|
4
|
Körner M, Müller P, Das H, Kraus F, Pfeuffer T, Spielhaupter S, Oeljeklaus S, Schülein-Völk C, Harper JW, Warscheid B, Buchberger A. p97/VCP is required for piecemeal autophagy of aggresomes. Nat Commun 2025; 16:4243. [PMID: 40335532 PMCID: PMC12059050 DOI: 10.1038/s41467-025-59556-x] [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: 07/12/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Metazoan cells adapt to the exhaustion of protein quality control (PQC) systems by sequestering aggregation-prone proteins in large, pericentriolar structures termed aggresomes. Defects in both aggresome formation and clearance affect proteostasis and have been linked to neurodegenerative diseases, but aggresome clearance pathways are still underexplored. Here we show that aggresomes comprising endogenous proteins are cleared via selective autophagy requiring the cargo receptor TAX1BP1. TAX1BP1 proximitomes reveal the presence of various PQC systems at aggresomes, including Hsp70 chaperones, the 26S proteasome, and the ubiquitin-selective unfoldase p97/VCP. While Hsp70 and p97/VCP with its cofactors UFD1-NPL4 and FAF1 play key roles in aggresome disassembly, the 26S proteasome is dispensable. We identify aggresomal client proteins that are degraded via different routes, in part in a p97/VCP-dependent manner via aggrephagy. Upon acute inhibition of p97/VCP, aggresomes fail to disintegrate and cannot be incorporated into autophagosomes despite the presence of factors critical for aggrephagosome formation, including p62/SQSTM1, TAX1BP1, and WIPI2. We conclude that the p97/VCP-mediated removal of ubiquitylated aggresomal clients is essential for the disintegration and subsequent piecemeal autophagy of aggresomes.
Collapse
Affiliation(s)
- Maria Körner
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Paul Müller
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Hirak Das
- Biocenter, Chair of Biochemistry II, University of Würzburg, Würzburg, Germany
| | - Felix Kraus
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Timo Pfeuffer
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Sven Spielhaupter
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Silke Oeljeklaus
- Biocenter, Chair of Biochemistry II, University of Würzburg, Würzburg, Germany
| | | | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Bettina Warscheid
- Biocenter, Chair of Biochemistry II, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
5
|
Vostal LE, Dahan NE, Reynolds MJ, Kronenberg LI, Kapoor TM. Structural insights into the coupling between VCP, an essential unfoldase, and a deubiquitinase. J Cell Biol 2025; 224:e202410148. [PMID: 40085114 PMCID: PMC11908451 DOI: 10.1083/jcb.202410148] [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: 10/24/2024] [Revised: 01/08/2025] [Accepted: 02/09/2025] [Indexed: 03/16/2025] Open
Abstract
Proteostasis involves degradation and recycling of proteins from organelles, membranes, and multiprotein complexes. These processes can depend on protein extraction and unfolding by the essential mechanoenzyme valosin-containing protein (VCP) and on ubiquitin chain remodeling by ubiquitin-specific proteases known as deubiquitinases (DUBs). How the activities of VCP and DUBs are coordinated is poorly understood. Here, we focus on the DUB VCPIP1, a VCP interactor required for post-mitotic Golgi and ER organization. We determine ∼3.3 Å cryogenic electron microscopy structures of VCP-VCPIP1 complexes in the absence of added nucleotide or the presence of an ATP analog. We find that up to 3 VCPIP1 protomers interact with the VCP hexamer to position VCPIP1's catalytic domain at the exit of VCP's central pore, poised to cleave ubiquitin following substrate unfolding. We observe competition between VCPIP1 and other cofactors for VCP binding and show that VCP stimulates VCPIP1's DUB activity. Together, our data suggest how the two enzyme activities can be coordinated to regulate proteostasis.
Collapse
Affiliation(s)
- Lauren E. Vostal
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Noa E. Dahan
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
- The David Rockefeller Graduate Program in Bioscience, The Rockefeller University, New York, NY, USA
| | - Matthew J. Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Lily I. Kronenberg
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
| | - Tarun M. Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
6
|
El Hebieshy AF, Wijfjes Z, Le Gall CM, Middelburg J, de Roode KE, Fennemann FL, Sluijter M, van Hall T, Dijkstra DJ, Trouw LA, van Dalen FJ, Rodgers Furones A, van der Schoot JMS, Derksen I, de Haard H, van der Woning B, Talavera Ormeño CMP, van Doodewaerd BR, Figdor CG, van der Heden van Noort GJ, Parren PWHI, Heskamp S, Ovaa H, Verdoes M, Scheeren FA. Site-directed multivalent conjugation of antibodies to ubiquitinated payloads. Nat Biomed Eng 2025:10.1038/s41551-024-01342-z. [PMID: 40204992 DOI: 10.1038/s41551-024-01342-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 12/20/2024] [Indexed: 04/11/2025]
Abstract
Antibody conjugates are the foundation of a wide range of diagnostic and therapeutic applications. Although many antibody-conjugation techniques are robust and efficient, obtaining homogeneous multimeric conjugation products remains challenging. Here we report a modular and versatile technique for the site-directed multivalent conjugation of antibodies via the small-protein ubiquitin. Specifically, multiple ubiquitin fusions with antibodies, antibody fragments, nanobodies, peptides or small molecules such as fluorescent dyes can be conjugated to antibodies and nanobodies within 30 min. The technique, which we named 'ubi-tagging', allowed us to efficiently generate a bispecific T-cell engager as well as nanobodies conjugated to dendritic-cell-targeted antigens that led to potent T-cell responses. Using both recombinant ubi-tagged proteins and synthetic ubiquitin derivatives allows for the iterative, site-directed and multivalent conjugation of antibodies and nanobodies to a plethora of molecular moieties.
Collapse
Affiliation(s)
- Angela F El Hebieshy
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Zacharias Wijfjes
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
- Institute for Chemical Immunology, Nijmegen, the Netherlands
| | - Camille M Le Gall
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jim Middelburg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Kim E de Roode
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Tagworks Pharmaceuticals, Nijmegen, the Netherlands
| | - Felix L Fennemann
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Douwe J Dijkstra
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Floris J van Dalen
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andrea Rodgers Furones
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Ian Derksen
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Cami M P Talavera Ormeño
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bjorn R van Doodewaerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Carl G Figdor
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Paul W H I Parren
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Huib Ovaa
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Martijn Verdoes
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
- Institute for Chemical Immunology, Nijmegen, the Netherlands.
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
7
|
Peck A, Dadi A, Yavarow Z, Alfano LN, Anderson D, Arkin MR, Chou TF, D'Ambrosio ES, Diaz-Manera J, Dudley JP, Elder AG, Ghoshal N, Hart CE, Hart MM, Huryn DM, Johnson AE, Jones KB, Kimonis V, Kiskinis E, Lee EB, Lloyd TE, Mapstone M, Martin A, Meyer H, Mozaffar T, Onyike CU, Pfeffer G, Pindon A, Raman M, Richard I, Rubinsztein DC, Schiava M, Schütz AK, Shen PS, Southworth DR, Staffaroni AM, Taralio-Gravovac M, Weihl CC, Yao Q, Ye Y, Peck N. 2024 VCP International Conference: Exploring multi-disciplinary approaches from basic science of valosin containing protein, an AAA+ ATPase protein, to the therapeutic advancement for VCP-associated multisystem proteinopathy. Neurobiol Dis 2025; 207:106861. [PMID: 40037468 PMCID: PMC11960434 DOI: 10.1016/j.nbd.2025.106861] [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: 01/25/2025] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
Valosin-containing protein (VCP/p97) is a ubiquitously expressed AAA+ ATPase associated with numerous protein-protein interactions and critical cellular functions including protein degradation and clearance, mitochondrial homeostasis, DNA repair and replication, cell cycle regulation, endoplasmic reticulum-associated degradation, and lysosomal functions including autophagy and apoptosis. Autosomal-dominant missense mutations in the VCP gene may result in VCP-associated multisystem proteinopathy (VCP-MSP), a rare degenerative disorder linked to heterogeneous phenotypes including inclusion body myopathy (IBM) with Paget's disease of bone (PDB) and frontotemporal dementia (FTD) or IBMPFD, amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), parkinsonism, Charcot-Marie Tooth disease (CMT), and spastic paraplegia. The complexity of VCP-MSP makes collaboration among stakeholders essential and necessitates a multi-disciplinary approach. The 2024 VCP International Conference was hosted at Caltech between February 22 and 25. Co-organized by Cure VCP Disease and Dr. Tsui-Fen Chou, the meeting aimed to center the patient as a research partner, harmonize diverse stakeholder engagement, and bridge the gap between basic and clinical neuroscience as it relates to VCP-MSP. Over 100 multi-disciplinary experts attended, ranging from basic scientists to clinicians to patient advocates. Attendees discussed genetics and clinical presentation, cellular and molecular mechanisms underlying disease, therapeutic approaches, and strategies for future VCP research. The conference included three roundtable discussions, 29 scientific presentations, 32 scientific posters, nine patient and caregiver posters, and a closing discussion forum. The following conference proceedings summarize these sessions, highlighting both the identified gaps in knowledge and the significant strides made towards understanding and treating VCP diseases.
Collapse
Affiliation(s)
- A Peck
- Cure VCP Disease, Warner Robins, GA, USA
| | - A Dadi
- Cure VCP Disease, Warner Robins, GA, USA
| | - Z Yavarow
- Cure VCP Disease, Warner Robins, GA, USA
| | - L N Alfano
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | | | - M R Arkin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - T F Chou
- Department of Biology, Caltech, Pasadena, CA, USA
| | - E S D'Ambrosio
- Nationwide Children's Hospital, Columbus, OH, USA; Department of Genetic and Cellular Medicine and Department of Neurology, UMass Chan Medical School, Worcester, MA, USA
| | - J Diaz-Manera
- The John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, Center for Life, Central Parkway, Newcastle, Upon Tyne, United Kingdom
| | - J P Dudley
- LaMontagne Center for Infectious Disease, University of Texas, Austin, TX, USA
| | - A G Elder
- Cure VCP Disease, Warner Robins, GA, USA
| | - N Ghoshal
- Departments of Neurology and Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - C E Hart
- Creyon Bio, San Diego, CA, USA; Lilly, Indianapolis, IN, USA
| | - M M Hart
- Cure VCP Disease, Warner Robins, GA, USA
| | - D M Huryn
- Department of Chemistry University of Pennsylvania, Philadelphia, PA, USA
| | - A E Johnson
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - K B Jones
- Department of Orthopaedics, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - V Kimonis
- Department of Pediatrics, University of California, Irvine, CA, USA; Department of Neurology, University of California, Irvine, CA, USA
| | - E Kiskinis
- The Ken & Ruth Davee Department of Neurology, Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - E B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - T E Lloyd
- Department of Neurology Baylor College of Medicine, Houston, TX, USA
| | - M Mapstone
- Department of Neurology, University of California, Irvine, CA, USA
| | - A Martin
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - H Meyer
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - T Mozaffar
- Department of Neurology, University of California, Irvine, CA, USA
| | - C U Onyike
- Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - G Pfeffer
- Department of Clinical Neurosciences, University of Calgary, Calgary, Canada; Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - A Pindon
- Cure VCP Disease, Warner Robins, GA, USA; Myhre Syndrome Foundation, Richardson, TX, USA
| | - M Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - I Richard
- Généthon, 91000 Evry, France; Université Paris-Saclay, Université Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry, France
| | - D C Rubinsztein
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, UK; UK Dementia Research Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - M Schiava
- The John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, Center for Life, Central Parkway, Newcastle, Upon Tyne, United Kingdom
| | - A K Schütz
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - P S Shen
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - D R Southworth
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
| | - A M Staffaroni
- UCSF Memory and Aging Center University of California San Francisco, CA, USA
| | - M Taralio-Gravovac
- Department of Biochemistry & Molecular Biology, University of Calgary, Alberta, Calgary, Canada
| | - C C Weihl
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Q Yao
- Department of Medicine Stony Brook University, Renaissance School of Medicine, Stony Brook, NY, USA
| | - Y Ye
- Laboratory of Molecular Biology, NIH, NIDDK, Bethesda, MD, USA
| | - N Peck
- Cure VCP Disease, Warner Robins, GA, USA.
| |
Collapse
|
8
|
McGirr T, Onar O, Jafarnejad SM. Dysregulated ribosome quality control in human diseases. FEBS J 2025; 292:936-959. [PMID: 38949989 PMCID: PMC11880988 DOI: 10.1111/febs.17217] [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] [Revised: 05/31/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Precise regulation of mRNA translation is of fundamental importance for maintaining homeostasis. Conversely, dysregulated general or transcript-specific translation, as well as abnormal translation events, have been linked to a multitude of diseases. However, driven by the misconception that the transient nature of mRNAs renders their abnormalities inconsequential, the importance of mechanisms that monitor the quality and fidelity of the translation process has been largely overlooked. In recent years, there has been a dramatic shift in this paradigm, evidenced by several seminal discoveries on the role of a key mechanism in monitoring the quality of mRNA translation - namely, Ribosome Quality Control (RQC) - in the maintenance of homeostasis and the prevention of diseases. Here, we will review recent advances in the field and emphasize the biological significance of the RQC mechanism, particularly its implications in human diseases.
Collapse
Affiliation(s)
- Tom McGirr
- Patrick G. Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Okan Onar
- Patrick G. Johnston Centre for Cancer ResearchQueen's University BelfastUK
- Department of Biology, Faculty of ScienceAnkara UniversityTurkey
| | | |
Collapse
|
9
|
Maduka AO, Manohar S, Foster MW, Silva GM. Localized K63 Ubiquitin Signaling Is Regulated by VCP/p97 During Oxidative Stress. Mol Cell Proteomics 2025; 24:100920. [PMID: 39880084 PMCID: PMC11894314 DOI: 10.1016/j.mcpro.2025.100920] [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: 06/10/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025] Open
Abstract
Under stress conditions, cells reprogram their molecular machineries to mitigate damage and promote survival. Ubiquitin signaling is globally increased during oxidative stress, controlling protein fate and supporting stress defenses at several subcellular compartments. However, the rules driving subcellular ubiquitin localization to promote concerted response mechanisms remain understudied. Here, we show that K63-linked polyubiquitin chains, known to promote proteasome-independent pathways, accumulate primarily in noncytosolic compartments during oxidative stress induced by sodium arsenite in mammalian cells. Our subcellular ubiquitin proteomic analyses of noncytosolic compartments expanded 2.5-fold the pool of proteins (2,494) and provided a comprehensive number of sites (10,157) known to be ubiquitinated during arsenite stress, suggesting their involvement in a myriad of cellular pathways. Moreover, subcellular proteome analyses revealed proteins that are recruited to noncytosolic compartments under stress, including a significant enrichment of helper ubiquitin-binding adaptors of the ATPase valosin-containing protein (VCP) that processes ubiquitinated substrates for downstream signaling. We further show that VCP recruitment to noncytosolic compartments under arsenite stress occurs in a ubiquitin-dependent manner mediated by its adaptor NPLOC4. Additionally, we show that VCP and NPLOC4 activities are critical to sustain low levels of noncytosolic K63-linked ubiquitin chains, supporting a cyclical model of ubiquitin conjugation and removal that is disrupted by reactive oxygen species. This work deepens our understanding of the role of localized ubiquitin and VCP signaling in the basic mechanisms of stress response and highlights new pathways and molecular players that are essential to reshape the composition and function of the human subcellular proteome under dynamic environments.
Collapse
Affiliation(s)
- Austin O Maduka
- Department of Biology, Duke University, Durham, North Carolina, USA
| | - Sandhya Manohar
- Department of Biology, Institute for Biochemistry, ETH Zürich, Zürich, Switzerland
| | - Matthew W Foster
- Proteomics and Metabolomics Core Facility, Duke University, Durham, North Carolina, USA
| | - Gustavo M Silva
- Department of Biology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
10
|
Ding R, Edwards TC, Goswami P, Wilson DJ, Dreis CD, Ye Y, Geraghty RJ, Chen L. p97 Inhibitors Possessing Antiviral Activity Against SARS-CoV-2 and Low Cytotoxicity. Pharmaceuticals (Basel) 2025; 18:131. [PMID: 39861192 PMCID: PMC11768289 DOI: 10.3390/ph18010131] [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: 12/04/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Background: p97 (also known as valosin-containing protein, VCP) is a member of the AAA+ ATPase family and is intimately associated with protein quality control and homeostasis regulation. Therefore, pharmaceutical inhibition of p97 has been actively pursued as an anticancer strategy. Recently, p97 has emerged as an important pro-viral host factor and p97 inhibitors are being evaluated as potential antiviral agents. Methods: We designed and synthesized novel p97 inhibitors based on the rearrangement of the central fused ring of our previously reported p97 inhibitors. These compounds were tested for inhibition of p97, cytotoxicity, and antiviral activity against SARS-CoV-2. Molecular docking was also performed on selected inhibitors to shed light on their binding modes. Results: Among these new p97 inhibitors, two compounds possess enhanced anti-p97 activity over their parent compounds. More significantly, these two inhibitors exhibit strong antiviral activity against SARS-CoV-2 at doses with no significant cytotoxicity. Molecular docking reveals no major change of the binding mode relative to that of their parent compounds, further supporting our design strategy. Conclusions: These compounds are structurally novel p97 inhibitors that display low toxicity and possess promising antiviral activity against SARS-CoV-2 and potentially other viruses. Further structural exploration is therefore justified and improved analogs will serve as useful tools for studying p97 as a promising host antiviral target.
Collapse
Affiliation(s)
- Rui Ding
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Tiffany C. Edwards
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Prithwish Goswami
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Daniel J. Wilson
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Christine D. Dreis
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Robert J. Geraghty
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Liqiang Chen
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| |
Collapse
|
11
|
Batra S, Vaquer-Alicea J, Valdez C, Taylor SP, Manon VA, Vega AR, Kashmer OM, Kolay S, Lemoff A, Cairns NJ, White CL, Diamond MI. VCP regulates early tau seed amplification via specific cofactors. Mol Neurodegener 2025; 20:2. [PMID: 39773263 PMCID: PMC11707990 DOI: 10.1186/s13024-024-00783-z] [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: 04/22/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Neurodegenerative tauopathies may progress based on seeding by pathological tau assemblies, whereby an aggregate is released from one cell, gains entry to an adjacent or connected cell, and serves as a specific template for its own replication in the cytoplasm. Seeding into the complex cytoplasmic milieu happens within hours, implying the existence of unknown factors that regulate this process. METHODS We used proximity labeling to identify proteins that control seed amplification within 5 h of seed exposure. We fused split-APEX2 to the C-terminus of tau repeat domain (RD) to reconstitute peroxidase activity 5 h after seeded intracellular tau aggregation. Valosin containing protein (VCP/p97) was the top hit. VCP harbors dominant mutations that underlie two neurodegenerative diseases, multisystem proteinopathy and vacuolar tauopathy, but its mechanistic role is unclear. We used immortalized cells and human neurons to study the effects of VCP on tau seeding. We exposed cells to fibrils or brain homogenates in cell culture media and measured effects on uptake and induction of intracellular tau aggregation following various genetic and pharmacological manipulations of VCP. RESULTS VCP knockdown reduced tau seeding. Chemical inhibitors had opposing effects on seeding in HEK293T tau biosensor cells and human neurons: ML-240 increased seeding efficiency, whereas NMS-873 decreased it. The inhibitors only functioned when administered within 8 h of seed exposure, indicating a role for VCP early in seed processing. We screened 30 VCP co-factors in HEK293T biosensor cells by genetic knockout or knockdown. Reduction of ATXN3, NSFL1C, UBE4B, NGLY1, and OTUB1 decreased tau seeding, as did NPLOC4, which also uniquely increased soluble tau levels. By contrast, reduction of FAF2 increased tau seeding. CONCLUSIONS Divergent effects on tau seeding of chemical inhibitors and cofactor reduction indicate that VCP regulates this process. This is consistent with a cytoplasmic processing complex centered on VCP that directs seeds acutely towards degradation vs. amplification.
Collapse
Affiliation(s)
- Sushobhna Batra
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Jaime Vaquer-Alicea
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Clarissa Valdez
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Skyler P Taylor
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Victor A Manon
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Anthony R Vega
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Omar M Kashmer
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Sourav Kolay
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Nigel J Cairns
- Department of Clinical and Biological Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Charles L White
- Department of Pathology, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States.
- Department of Neurology, Dallas, United States.
| |
Collapse
|
12
|
Cui YH, Wei J, Fan H, Li W, Zhao L, Wilkinson E, Peterson J, Xie L, Zou Z, Yang S, Applebaum MA, Kline J, Chen J, He C, He YY. Targeting DTX2/UFD1-mediated FTO degradation to regulate antitumor immunity. Proc Natl Acad Sci U S A 2024; 121:e2407910121. [PMID: 39661064 DOI: 10.1073/pnas.2407910121] [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: 04/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Here, we show that vitamin E succinate (VES) acts as a degrader for the m6A RNA demethylase fat mass and obesity-associated protein (FTO), thus suppressing tumor growth and resistance to immunotherapy. FTO is ubiquitinated by its E3 ligase DTX2, followed by UFD1 recruitment and subsequent degradation in the proteasome. VES binds to FTO and DTX2, leading to enhanced FTO-DTX2 interaction, FTO ubiquitination, and degradation in FTO-dependent tumor cells. VES suppressed tumor growth and enhanced antitumor immunity and response to immunotherapy in vivo in mouse models. Genetic FTO knockdown or VES treatment increased m6A methylation in the LIF (Leukemia Inhibitory Factor) gene and decreased LIF mRNA decay, and thus sensitized melanoma cells to T cell-mediated cytotoxicity. Taken together, our findings reveal the underlying molecular mechanism for FTO protein degradation and identify a dietary degrader for FTO that inhibits tumor growth and overcomes immunotherapy resistance.
Collapse
Affiliation(s)
- Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Jiangbo Wei
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Hao Fan
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Wenlong Li
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Lijie Zhao
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Emma Wilkinson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637
| | - Jack Peterson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- The College, University of Chicago, Chicago, IL 60637
| | - Lishi Xie
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Zhongyu Zou
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Mark A Applebaum
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637
| | - Justin Kline
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Jing Chen
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Chuan He
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
- HHMI, University of Chicago, Chicago, IL 60637
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
13
|
Ren K, Luan Y, Sun Y, Huang S, Zhang S, Yang Y, Jin Y, Chen X. NPLOC4 aggravates heart failure by regulating ROS and mitochondrial function. Int Immunopharmacol 2024; 142:113199. [PMID: 39332095 DOI: 10.1016/j.intimp.2024.113199] [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: 07/05/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 09/29/2024]
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality worldwide, necessitating the discovery of new therapeutic targets. NPLOC4 is known as an endoplasmic reticulum protein involved in protein degradation and cellular stress responses. Herein, NPLOC4 was investigated for its role in HF using a transverse aortic constriction (TAC) mouse model and an Angiotensin II (Ang II)-induced H9c2 cardiomyocyte model. Transcriptomic analysis revealed NPLOC4 upregulation in HF. NPLOC4 knockdown in the TAC model inhibited HF progression, as evidenced by reduced cardiac hypertrophy and fibrosis. Subsequent knockdown experiments showed the relievement in heart failure phenotypes, reduced reactive oxygen species (ROS) levels and enhanced mitochondrial function caused by NPLOC4 depletion in Ang II-induced H9c2 cells. STRING analysis predicted ERO1α as a potential NPLOC4 interactor, with further studies identifying that NPLOC4 knockdown increases ERO1α expression and disrupts mitochondria-associated membranes (MAMs). Additionally, NPLOC4 knockdown modulated the β-catenin/GSK3β pathway, enhancing mitochondrial dynamics and mitophagy. These findings suggest NPLOC4 as a promising therapeutic target for HF.
Collapse
Affiliation(s)
- Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yi Luan
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yuanyuan Sun
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siyuan Huang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shuwei Zhang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Yang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Xing Chen
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
14
|
Masuda R, Yoshikawa M, Moriuchi R, Oba Y, Dohra H, Kimura Y. Elucidation of Ubiquitin-Related Functions via an Ubiquitin Overexpression Approach. Cells 2024; 13:2011. [PMID: 39682759 PMCID: PMC11639773 DOI: 10.3390/cells13232011] [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: 11/11/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
To identify new ubiquitin-related functions using yeast, we searched for mutants conferring a temperature-sensitivity phenotype that could be rescued through ubiquitin overexpression. Screening of mutants using this overexpression strategy identified SPC2, which encodes a subunit of the endoplasmic reticulum (ER) signal peptidase complex (SPC). Ubiquitin overexpression rescued a high-temperature sensitivity of spc2 deletion mutant, suggesting that ubiquitin could compensate for Spc2 loss-of-function at high temperatures. The double mutant of Spc2 and Hrd1, an ER E3 ubiquitin ligase, showed a synergistic growth defect at higher temperatures. A weak genetic interaction was also observed between spc2Δ and cdc48-3 mutation. The results suggest a close functional relationship between SPC and the ubiquitin-proteasome system in yeast and further provide proof-of-principle for this ubiquitin overexpression approach to identify novel ubiquitin-related genes and associated cellular processes.
Collapse
Affiliation(s)
- Ryo Masuda
- Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan (H.D.)
| | - Munetaka Yoshikawa
- Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan (H.D.)
| | - Ryota Moriuchi
- Shizuoka Instrumental Analysis Center, Shizuoka University, Shizuoka 422-8529, Japan;
| | - Yumiko Oba
- Department of Agriculture, Shizuoka University, Shizuoka 422-8529, Japan;
| | - Hideo Dohra
- Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan (H.D.)
- Shizuoka Instrumental Analysis Center, Shizuoka University, Shizuoka 422-8529, Japan;
- Research Institute of Green Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan
| | - Yoko Kimura
- Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan (H.D.)
- Department of Agriculture, Shizuoka University, Shizuoka 422-8529, Japan;
| |
Collapse
|
15
|
Lange SM, McFarland MR, Lamoliatte F, Carroll T, Krshnan L, Pérez-Ràfols A, Kwasna D, Shen L, Wallace I, Cole I, Armstrong LA, Knebel A, Johnson C, De Cesare V, Kulathu Y. VCP/p97-associated proteins are binders and debranching enzymes of K48-K63-branched ubiquitin chains. Nat Struct Mol Biol 2024; 31:1872-1887. [PMID: 38977901 PMCID: PMC11638074 DOI: 10.1038/s41594-024-01354-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: 09/25/2023] [Accepted: 06/13/2024] [Indexed: 07/10/2024]
Abstract
Branched ubiquitin (Ub) chains constitute a sizable fraction of Ub polymers in human cells. Despite their abundance, our understanding of branched Ub function in cell signaling has been stunted by the absence of accessible methods and tools. Here we identify cellular branched-chain-specific binding proteins and devise approaches to probe K48-K63-branched Ub function. We establish a method to monitor cleavage of linkages within complex Ub chains and unveil ATXN3 and MINDY as debranching enzymes. We engineer a K48-K63 branch-specific nanobody and reveal the molecular basis of its specificity in crystal structures of nanobody-branched Ub chain complexes. Using this nanobody, we detect increased K48-K63-Ub branching following valosin-containing protein (VCP)/p97 inhibition and after DNA damage. Together with our discovery that multiple VCP/p97-associated proteins bind to or debranch K48-K63-linked Ub, these results suggest a function for K48-K63-branched chains in VCP/p97-related processes.
Collapse
Affiliation(s)
- Sven M Lange
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| | - Matthew R McFarland
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Frederic Lamoliatte
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Thomas Carroll
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Logesvaran Krshnan
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Anna Pérez-Ràfols
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Dominika Kwasna
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
- Malopolska Centre of Biotechnology (MCB), Jagiellonian University, Krakow, Poland
| | - Linnan Shen
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Iona Wallace
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Isobel Cole
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Lee A Armstrong
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Axel Knebel
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Clare Johnson
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Virginia De Cesare
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Yogesh Kulathu
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK.
| |
Collapse
|
16
|
Ingersoll AJ, McCloud DM, Hu JY, Rape M. Dynamic regulation of the oxidative stress response by the E3 ligase TRIP12. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625235. [PMID: 39651249 PMCID: PMC11623662 DOI: 10.1101/2024.11.25.625235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The oxidative stress response is centered on the transcription factor NRF2 and protects cells from reactive oxygen species (ROS). While ROS inhibit the E3 ligase CUL3 KEAP1 to stabilize NRF2 and elicit antioxidant gene expression, cells recovering from stress must rapidly reactivate CUL3 KEAP1 to prevent reductive stress and oxeiptosis-dependent cell death. How cells restore efficient NRF2-degradation upon ROS clearance remains poorly understood. Here, we identify TRIP12, an E3 ligase dysregulated in Clark-Baraitser Syndrome and Parkinson's Disease, as a component of the oxidative stress response. TRIP12 is a ubiquitin chain elongation factor that cooperates with CUL3 KEAP1 to ensure robust NRF2 degradation. In this manner, TRIP12 accelerates stress response silencing as ROS are being cleared, but limits NRF2 activation during stress. The need for dynamic control of NRF2-degradation therefore comes at the cost of diminished stress signaling, suggesting that TRIP12 inhibition could be used to treat degenerative pathologies characterized by ROS accumulation.
Collapse
|
17
|
Findlay AR. Dominantly inherited muscle disorders: understanding their complexity and exploring therapeutic approaches. Dis Model Mech 2024; 17:dmm050720. [PMID: 39501809 PMCID: PMC11574355 DOI: 10.1242/dmm.050720] [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] [Indexed: 11/21/2024] Open
Abstract
Treatments for disabling and life-threatening hereditary muscle disorders are finally close to becoming a reality. Research has thus far focused primarily on recessive forms of muscle disease. The gene replacement strategies that are commonly employed for recessive, loss-of-function disorders are not readily translatable to most dominant myopathies owing to the presence of a normal chromosome in each nucleus, hindering the development of novel treatments for these dominant disorders. This is largely due to their complex, heterogeneous disease mechanisms that require unique therapeutic approaches. However, as viral and RNA interference-based therapies enter clinical use, key tools are now in place to develop treatments for dominantly inherited disorders of muscle. This article will review what is known about dominantly inherited disorders of muscle, specifically their genetic basis, how mutations lead to disease, and the pathomechanistic implications for therapeutic approaches.
Collapse
Affiliation(s)
- Andrew R Findlay
- Washington University Saint Louis, Neuromuscular Disease Center, 660 S. Euclid Ave., St Louis, MO 63110, USA
| |
Collapse
|
18
|
Giong HK, Hyeon SJ, Lee JG, Cho HJ, Park U, Stein TD, Lee J, Yu K, Ryu H, Lee JS. Tau accumulation is cleared by the induced expression of VCP via autophagy. Acta Neuropathol 2024; 148:46. [PMID: 39316141 PMCID: PMC11422276 DOI: 10.1007/s00401-024-02804-z] [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: 03/15/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/25/2024]
Abstract
Tauopathy, including frontotemporal lobar dementia and Alzheimer's disease, describes a class of neurodegenerative diseases characterized by the aberrant accumulation of Tau protein due to defects in proteostasis. Upon generating and characterizing a stable transgenic zebrafish that expresses the human TAUP301L mutant in a neuron-specific manner, we found that accumulating Tau protein was efficiently cleared via an enhanced autophagy activity despite constant Tau mRNA expression; apparent tauopathy-like phenotypes were revealed only when the autophagy was genetically or chemically inhibited. We performed RNA-seq analysis, genetic knockdown, and rescue experiments with clinically relevant point mutations of valosin-containing protein (VCP), and showed that induced expression of VCP, an essential cytosolic chaperone for the protein quality system, was a key factor for Tau degradation via its facilitation of the autophagy flux. This novel function of VCP in Tau clearance was further confirmed in a tauopathy mouse model where VCP overexpression significantly decreased the level of phosphorylated and oligomeric/aggregate Tau and rescued Tau-induced cognitive behavioral phenotypes, which were reversed when the autophagy was blocked. Importantly, VCP expression in the brains of human Alzheimer's disease patients was severely downregulated, consistent with its proposed role in Tau clearance. Taken together, these results suggest that enhancing the expression and activity of VCP in a spatiotemporal manner to facilitate the autophagy pathway is a potential therapeutic approach for treating tauopathy.
Collapse
Affiliation(s)
- Hoi-Khoanh Giong
- Microbiome Convergence Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Greenwood Genetic Center, Greenwood, SC, 29646, USA
| | - Seung Jae Hyeon
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jae-Geun Lee
- Microbiome Convergence Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Hyun-Ju Cho
- Microbiome Convergence Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Uiyeol Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Thor D Stein
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Junghee Lee
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Kweon Yu
- Disease Target Structure Research Centre, KRIBB, Daejeon, 34141, Republic of Korea
- KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hoon Ryu
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Jeong-Soo Lee
- Microbiome Convergence Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea.
- Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| |
Collapse
|
19
|
Vostal LE, Dahan NE, Zhang W, Reynolds MJ, Chait BT, Kapoor TM. Distinct modes of coupling between VCP, an essential unfoldase, and deubiquitinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.08.611915. [PMID: 39314330 PMCID: PMC11418955 DOI: 10.1101/2024.09.08.611915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Errors in proteostasis, which requires regulated degradation and recycling of diverse proteins, are linked to aging, cancer and neurodegenerative disease (1). In particular, recycling proteins from multiprotein complexes, organelles and membranes is initiated by ubiquitylation, extraction and unfolding by the essential mechanoenzyme VCP (2-4), and ubiquitin removal by deubiquitinases (DUBs), a class of ∼100 ubiquitin-specific proteases in humans (5, 6). As VCP's substrate recognition requires ubiquitylation, the removal of ubiquitins from substrates for recycling must follow extraction and unfolding. How the activities of VCP and different DUBs are coordinated for protein recycling or other fates is unclear. Here, we employ a photochemistry-based approach to profile proteome-wide domain-specific VCP interactions in living cells (7). We identify DUBs that bind near the entry, exit, or both sites of VCP's central pore, the channel for ATP-dependent substrate translocation (8-10). From this set of DUBs, we focus on VCPIP1, required for organelle assembly and DNA repair (11-13), that our chemical proteomics workflow indicates binds the central pore's entry and exit sites. We determine a ∼3Å cryo-EM structure of the VCP-VCPIP1 complex and find up to 3 VCPIP1 protomers interact with the VCP hexamer. VCPIP1's UBX-L domain binds VCP's N-domain in a 'down' conformation, linked to VCP's ADP-bound state (2, 14), and the deubiquitinase domain is positioned at the central pore's exit site, poised to remove ubiquitin following substrate unfolding. We find that VCP stimulates VCPIP1's DUB activity and use mutagenesis and single-molecule mass photometry assays to test the structural model. Together, our data suggest that DUBs bind VCP at distinct sites and reveal how the two enzyme activities can be coordinated to achieve specific downstream outcomes for ubiquitylated proteins.
Collapse
|
20
|
Wang J, Chistov G, Zhang J, Huntington B, Salem I, Sandholu A, Arold ST. P-NADs: PUX-based NAnobody degraders for ubiquitin-independent degradation of target proteins. Heliyon 2024; 10:e34487. [PMID: 39130484 PMCID: PMC11315185 DOI: 10.1016/j.heliyon.2024.e34487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Targeted protein degradation (TPD) allows cells to maintain a functional proteome and to rapidly adapt to changing conditions. Methods that repurpose TPD for the deactivation of specific proteins have demonstrated significant potential in therapeutic and research applications. Most of these methods are based on proteolysis targeting chimaeras (PROTACs) which link the protein target to an E3 ubiquitin ligase, resulting in the ubiquitin-based degradation of the target protein. In this study, we introduce a method for ubiquitin-independent TPD based on nanobody-conjugated plant ubiquitin regulatory X domain-containing (PUX) adaptor proteins. We show that the PUX-based NAnobody Degraders (P-NADs) can unfold a target protein through the Arabidopsis and human orthologues of the CDC48 unfoldase without the need for ubiquitination or initiating motifs. We demonstrate that P-NAD plasmids can be transfected into a human cell line, where the produced P-NADs use the endogenous CDC48 machinery for ubiquitin-independent TPD of a 143 kDa multidomain protein. Thus, P-NADs pave the road for ubiquitin-independent therapeutic TPD approaches. In addition, the modular P-NAD design combined with in vitro and cellular assays provide a versatile platform for elucidating functional aspects of CDC48-based TPD in plants and animals.
Collapse
Affiliation(s)
- Jun Wang
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | | | - Junrui Zhang
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Brandon Huntington
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Israa Salem
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Anandsukeerthi Sandholu
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Stefan T. Arold
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
21
|
Kim JW, Bae JH, Go GY, Lee JR, Jeong Y, Kim JY, Kim TH, Kim YK, Han JW, Oh JE, Hahn MJ, Kang JS, Bae GU. Epsti1 Regulates the Inflammatory Stage of Early Muscle Regeneration through STAT1-VCP Interaction. Int J Biol Sci 2024; 20:3530-3543. [PMID: 38993551 PMCID: PMC11234217 DOI: 10.7150/ijbs.94675] [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: 01/25/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
During muscle regeneration, interferon-gamma (IFN-γ) coordinates inflammatory responses critical for activation of quiescent muscle stem cells upon injury via the Janus kinase (JAK) - signal transducer and activator of transcription 1 (STAT1) pathway. Dysregulation of JAK-STAT1 signaling results in impaired muscle regeneration, leading to muscle dysfunction or muscle atrophy. Until now, the underlying molecular mechanism of how JAK-STAT1 signaling resolves during muscle regeneration remains largely elusive. Here, we demonstrate that epithelial-stromal interaction 1 (Epsti1), an interferon response gene, has a crucial role in regulating the IFN-γ-JAK-STAT1 signaling at early stage of muscle regeneration. Epsti1-deficient mice exhibit impaired muscle regeneration with elevated inflammation response. In addition, Epsti1-deficient myoblasts display aberrant interferon responses. Epsti1 interacts with valosin-containing protein (VCP) and mediates the proteasomal degradation of IFN-γ-activated STAT1, likely contributing to dampening STAT1-mediated inflammation. In line with the notion, mice lacking Epsti1 exhibit exacerbated muscle atrophy accompanied by increased inflammatory response in cancer cachexia model. Our study suggests a crucial function of Epsti1 in the resolution of IFN-γ-JAK-STAT1 signaling through interaction with VCP which provides insights into the unexplored mechanism of crosstalk between inflammatory response and muscle regeneration.
Collapse
Affiliation(s)
- Jee Won Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Ju-Hyeon Bae
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
| | - Ga-Yeon Go
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Jae-Rin Lee
- Cell and Gene Therapy Products Division, National Institute of Food and Drug Safety Evaluation, Cheongju 28159, South Korea
| | - Yideul Jeong
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| | - Jun-Young Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Tae Hyun Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Yong Kee Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Jeung-Whan Han
- Research Center for Epigenome Regulation, School of Pharmacy, Department of Biochemistry and Molecular Biology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Ji-Eun Oh
- Department of Biomedical Laboratory Science, Far East University, 76-32 Daehakgil, Gamgok-myeon, Eumseong-gun, Chungbuk-do, 27601, Korea
| | - Myong-Joon Hahn
- Research Center for Epigenome Regulation, School of Pharmacy, Department of Biochemistry and Molecular Biology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| | - Gyu-Un Bae
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| |
Collapse
|
22
|
Maduka AO, Manohar S, Foster MW, Silva GM. Localized K63 ubiquitin signaling is regulated by VCP/p97 during oxidative stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.598218. [PMID: 38948861 PMCID: PMC11213022 DOI: 10.1101/2024.06.20.598218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Under stress conditions, cells reprogram their molecular machineries to mitigate damage and promote survival. Ubiquitin signaling is globally increased during oxidative stress, controlling protein fate and supporting stress defenses at several subcellular compartments. However, the rules driving subcellular ubiquitin localization to promote these concerted response mechanisms remain understudied. Here, we show that K63-linked ubiquitin chains, known to promote proteasome-independent pathways, accumulate primarily in non-cytosolic compartments during oxidative stress induced by sodium arsenite in mammalian cells. Our subcellular ubiquitin proteomic analyses of non-cytosolic compartments expanded 10-fold the pool of proteins known to be ubiquitinated during arsenite stress (2,046) and revealed their involvement in pathways related to immune signaling and translation control. Moreover, subcellular proteome analyses revealed proteins that are recruited to non-cytosolic compartments under stress, including a significant enrichment of helper ubiquitin-binding adaptors of the ATPase VCP that processes ubiquitinated substrates for downstream signaling. We further show that VCP recruitment to non-cytosolic compartments under arsenite stress occurs in a ubiquitin-dependent manner mediated by its adaptor NPLOC4. Additionally, we show that VCP and NPLOC4 activities are critical to sustain low levels of non-cytosolic K63-linked ubiquitin chains, supporting a cyclical model of ubiquitin conjugation and removal that is disrupted by cellular exposure to reactive oxygen species. This work deepens our understanding of the role of localized ubiquitin and VCP signaling in the basic mechanisms of stress response and highlights new pathways and molecular players that are essential to reshape the composition and function of the human subcellular proteome under dynamic environments.
Collapse
Affiliation(s)
| | - Sandhya Manohar
- Institute for Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Matthew W. Foster
- Proteomics and Metabolomics Core Facility, Duke University, Durham, NC, 27708, USA
| | | |
Collapse
|
23
|
Jones NH, Liu Q, Urnavicius L, Dahan NE, Vostal LE, Kapoor TM. Allosteric activation of VCP, an AAA unfoldase, by small molecule mimicry. Proc Natl Acad Sci U S A 2024; 121:e2316892121. [PMID: 38833472 PMCID: PMC11181084 DOI: 10.1073/pnas.2316892121] [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: 10/02/2023] [Accepted: 04/17/2024] [Indexed: 06/06/2024] Open
Abstract
The loss of function of AAA (ATPases associated with diverse cellular activities) mechanoenzymes has been linked to diseases, and small molecules that activate these proteins can be powerful tools to probe mechanisms and test therapeutic hypotheses. Unlike chemical inhibitors that can bind a single conformational state to block enzyme function, activator binding must be permissive to different conformational states needed for mechanochemistry. However, we do not know how AAA proteins can be activated by small molecules. Here, we focus on valosin-containing protein (VCP)/p97, an AAA unfoldase whose loss of function has been linked to protein aggregation-based disorders, to identify druggable sites for chemical activators. We identified VCP ATPase Activator 1 (VAA1), a compound that dose-dependently stimulates VCP ATPase activity up to ~threefold. Our cryo-EM studies resulted in structures (ranging from ~2.9 to 3.7 Å-resolution) of VCP in apo and ADP-bound states and revealed that VAA1 binds an allosteric pocket near the C-terminus in both states. Engineered mutations in the VAA1-binding site confer resistance to VAA1, and furthermore, modulate VCP activity. Mutation of a phenylalanine residue in the VCP C-terminal tail that can occupy the VAA1 binding site also stimulates ATPase activity, suggesting that VAA1 acts by mimicking this interaction. Together, our findings uncover a druggable allosteric site and a mechanism of enzyme regulation that can be tuned through small molecule mimicry.
Collapse
Affiliation(s)
- Natalie H. Jones
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY10065
- Tri-Institutional PhD Program in Chemical Biology, New York, NY10065
| | - Qiwen Liu
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY10065
| | - Linas Urnavicius
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY10065
| | - Noa E. Dahan
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY10065
- The David Rockefeller Graduate Program in Bioscience, The Rockefeller University, New York, NY10065
| | - Lauren E. Vostal
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY10065
- Tri-Institutional PhD Program in Chemical Biology, New York, NY10065
| | - Tarun M. Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY10065
| |
Collapse
|
24
|
Phan JM, Creekmore BC, Nguyen AT, Bershadskaya DD, Darwich NF, Mann CN, Lee EB. VCP activator reverses nuclear proteostasis defects and enhances TDP-43 aggregate clearance in multisystem proteinopathy models. J Clin Invest 2024; 134:e169039. [PMID: 38787785 PMCID: PMC11257039 DOI: 10.1172/jci169039] [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: 01/20/2023] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Pathogenic variants in valosin-containing protein (VCP) cause multisystem proteinopathy (MSP), a disease characterized by multiple clinical phenotypes including inclusion body myopathy, Paget's disease of the bone, and frontotemporal dementia (FTD). How such diverse phenotypes are driven by pathogenic VCP variants is not known. We found that these diseases exhibit a common pathologic feature: ubiquitinated intranuclear inclusions affecting myocytes, osteoclasts, and neurons. Moreover, knock-in cell lines harboring MSP variants show a reduction in nuclear VCP. Given that MSP is associated with neuronal intranuclear inclusions comprised of TDP-43 protein, we developed a cellular model whereby proteostatic stress results in the formation of insoluble intranuclear TDP-43 aggregates. Consistent with a loss of nuclear VCP function, cells harboring MSP variants or cells treated with VCP inhibitor exhibited decreased clearance of insoluble intranuclear TDP-43 aggregates. Moreover, we identified 4 compounds that activate VCP primarily by increasing D2 ATPase activity, where pharmacologic VCP activation appears to enhance clearance of insoluble intranuclear TDP-43 aggregate. Our findings suggest that VCP function is important for nuclear protein homeostasis, that impaired nuclear proteostasis may contribute to MSP, and that VCP activation may be a potential therapeutic by virtue of enhancing the clearance of intranuclear protein aggregates.
Collapse
|
25
|
Pontifex CS, Zaman M, Fanganiello RD, Shutt TE, Pfeffer G. Valosin-Containing Protein (VCP): A Review of Its Diverse Molecular Functions and Clinical Phenotypes. Int J Mol Sci 2024; 25:5633. [PMID: 38891822 PMCID: PMC11172259 DOI: 10.3390/ijms25115633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
In this review we examine the functionally diverse ATPase associated with various cellular activities (AAA-ATPase), valosin-containing protein (VCP/p97), its molecular functions, the mutational landscape of VCP and the phenotypic manifestation of VCP disease. VCP is crucial to a multitude of cellular functions including protein quality control, endoplasmic reticulum-associated degradation (ERAD), autophagy, mitophagy, lysophagy, stress granule formation and clearance, DNA replication and mitosis, DNA damage response including nucleotide excision repair, ATM- and ATR-mediated damage response, homologous repair and non-homologous end joining. VCP variants cause multisystem proteinopathy, and pathology can arise in several tissue types such as skeletal muscle, bone, brain, motor neurons, sensory neurons and possibly cardiac muscle, with the disease course being challenging to predict.
Collapse
Affiliation(s)
- Carly S. Pontifex
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (C.S.P.); (M.Z.); (T.E.S.)
| | - Mashiat Zaman
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (C.S.P.); (M.Z.); (T.E.S.)
- Alberta Child Health Research Institute, Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Timothy E. Shutt
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (C.S.P.); (M.Z.); (T.E.S.)
- Alberta Child Health Research Institute, Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (C.S.P.); (M.Z.); (T.E.S.)
- Alberta Child Health Research Institute, Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Heritage Medical Research Building 155, 3330 Hospital Dr NW, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
26
|
Batra S, Vaquer-Alicea JI, Valdez C, Taylor SP, Manon VA, Vega AR, Kashmer OM, Kolay S, Lemoff A, Cairns NJ, White CL, Diamond MI. VCP regulates early tau seed amplification via specific cofactors. RESEARCH SQUARE 2024:rs.3.rs-4307848. [PMID: 38826306 PMCID: PMC11142303 DOI: 10.21203/rs.3.rs-4307848/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Neurodegenerative tauopathies may progress based on seeding by pathological tau assemblies, whereby an aggregate is released from one cell, gains entry to an adjacent or connected cell, and serves as a specific template for its own replication in the cytoplasm. In vitro seeding reactions typically take days, yet seeding into the complex cytoplasmic milieu happens within hours, implicating a machinery with unknown players that controls this process in the acute phase. Methods We used proximity labeling to identify factors that control seed amplification within 5h of seed exposure. We fused split-APEX2 to the C-terminus of tau repeat domain (RD) to reconstitute peroxidase activity 5h after seeded intracellular tau aggregation. Valosin containing protein (VCP/p97) was the top hit. VCP harbors dominant mutations that underlie two neurodegenerative diseases, multisystem proteinopathy and vacuolar tauopathy, but its mechanistic role is unclear. We used immortalized cells and human neurons to study the effects of VCP on tau seeding. We exposed cells to fibrils or brain homogenates in cell culture media and measured effects on uptake and induction of intracellular tau aggregation following various genetic and chemical manipulations of VCP. Results VCP knockdown reduced tau seeding. Chemical inhibitors had opposing effects on aggregation in HEK293T tau biosensor cells and human neurons alike: ML-240 increased seeding efficiency, whereas NMS-873 decreased it. The inhibitors were effective only when administered within 8h of seed exposure, indicating a role for VCP early in seed processing. We screened 30 VCP co-factors in HEK293T biosensor cells by genetic knockout or knockdown. Reduction of ATXN3, NSFL1C, UBE4B, NGLY1, and OTUB1 decreased tau seeding, as did NPLOC4, which also uniquely increased soluble tau levels. By contrast, reduction of FAF2 increased tau seeding. Conclusions Divergent effects on tau seeding of chemical inhibitors and cofactor reduction indicate that VCP regulates this process. This is consistent with a dedicated cytoplasmic processing complex based on VCP that directs seeds acutely towards degradation vs. amplification.
Collapse
Affiliation(s)
- Sushobhna Batra
- UT Southwestern: The University of Texas Southwestern Medical Center
| | | | - Clarissa Valdez
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Skyler P Taylor
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Victor A Manon
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Anthony R Vega
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Omar M Kashmer
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Sourav Kolay
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Andrew Lemoff
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Nigel J Cairns
- University of Exeter Faculty of Health and Life Sciences
| | - Charles L White
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Marc I Diamond
- UT Southwestern: The University of Texas Southwestern Medical Center
| |
Collapse
|
27
|
Krishnamoorthy V, Foglizzo M, Dilley RL, Wu A, Datta A, Dutta P, Campbell LJ, Degtjarik O, Musgrove LJ, Calabrese AN, Zeqiraj E, Greenberg RA. The SPATA5-SPATA5L1 ATPase complex directs replisome proteostasis to ensure genome integrity. Cell 2024; 187:2250-2268.e31. [PMID: 38554706 PMCID: PMC11055677 DOI: 10.1016/j.cell.2024.03.002] [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: 02/02/2023] [Revised: 12/27/2023] [Accepted: 03/02/2024] [Indexed: 04/02/2024]
Abstract
Ubiquitin-dependent unfolding of the CMG helicase by VCP/p97 is required to terminate DNA replication. Other replisome components are not processed in the same fashion, suggesting that additional mechanisms underlie replication protein turnover. Here, we identify replisome factor interactions with a protein complex composed of AAA+ ATPases SPATA5-SPATA5L1 together with heterodimeric partners C1orf109-CINP (55LCC). An integrative structural biology approach revealed a molecular architecture of SPATA5-SPATA5L1 N-terminal domains interacting with C1orf109-CINP to form a funnel-like structure above a cylindrically shaped ATPase motor. Deficiency in the 55LCC complex elicited ubiquitin-independent proteotoxicity, replication stress, and severe chromosome instability. 55LCC showed ATPase activity that was specifically enhanced by replication fork DNA and was coupled to cysteine protease-dependent cleavage of replisome substrates in response to replication fork damage. These findings define 55LCC-mediated proteostasis as critical for replication fork progression and genome stability and provide a rationale for pathogenic variants seen in associated human neurodevelopmental disorders.
Collapse
Affiliation(s)
- Vidhya Krishnamoorthy
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Martina Foglizzo
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Robert L Dilley
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA.
| | - Angela Wu
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Arindam Datta
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Parul Dutta
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Lisa J Campbell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Oksana Degtjarik
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Laura J Musgrove
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Antonio N Calabrese
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Elton Zeqiraj
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| | - Roger A Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA.
| |
Collapse
|
28
|
Li H, Ji Z, Paulo JA, Gygi SP, Rapoport TA. Bidirectional substrate shuttling between the 26S proteasome and the Cdc48 ATPase promotes protein degradation. Mol Cell 2024; 84:1290-1303.e7. [PMID: 38401542 PMCID: PMC11995671 DOI: 10.1016/j.molcel.2024.01.029] [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: 02/01/2023] [Revised: 12/12/2023] [Accepted: 01/31/2024] [Indexed: 02/26/2024]
Abstract
Most eukaryotic proteins are degraded by the 26S proteasome after modification with a polyubiquitin chain. Substrates lacking unstructured segments cannot be degraded directly and require prior unfolding by the Cdc48 ATPase (p97 or VCP in mammals) in complex with its ubiquitin-binding partner Ufd1-Npl4 (UN). Here, we use purified yeast components to reconstitute Cdc48-dependent degradation of well-folded model substrates by the proteasome. We show that a minimal system consists of the 26S proteasome, the Cdc48-UN ATPase complex, the proteasome cofactor Rad23, and the Cdc48 cofactors Ubx5 and Shp1. Rad23 and Ubx5 stimulate polyubiquitin binding to the 26S proteasome and the Cdc48-UN complex, respectively, allowing these machines to compete for substrates before and after their unfolding. Shp1 stimulates protein unfolding by the Cdc48-UN complex rather than substrate recruitment. Experiments in yeast cells confirm that many proteins undergo bidirectional substrate shuttling between the 26S proteasome and Cdc48 ATPase before being degraded.
Collapse
Affiliation(s)
- Hao Li
- Howard Hughes Medical Institute and Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Zhejian Ji
- Howard Hughes Medical Institute and Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Tom A Rapoport
- Howard Hughes Medical Institute and Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Mirsanaye AS, Hoffmann S, Weisser M, Mund A, Lopez Mendez B, Typas D, van den Boom J, Benedict B, Hendriks IA, Nielsen ML, Meyer H, Duxin JP, Montoya G, Mailand N. VCF1 is a p97/VCP cofactor promoting recognition of ubiquitylated p97-UFD1-NPL4 substrates. Nat Commun 2024; 15:2459. [PMID: 38503733 PMCID: PMC10950897 DOI: 10.1038/s41467-024-46760-4] [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: 07/20/2023] [Accepted: 03/07/2024] [Indexed: 03/21/2024] Open
Abstract
The hexameric AAA+ ATPase p97/VCP functions as an essential mediator of ubiquitin-dependent cellular processes, extracting ubiquitylated proteins from macromolecular complexes or membranes by catalyzing their unfolding. p97 is directed to ubiquitylated client proteins via multiple cofactors, most of which interact with the p97 N-domain. Here, we discover that FAM104A, a protein of unknown function also named VCF1 (VCP/p97 nuclear Cofactor Family member 1), acts as a p97 cofactor in human cells. Detailed structure-function studies reveal that VCF1 directly binds p97 via a conserved α-helical motif that recognizes the p97 N-domain with unusually high affinity, exceeding that of other cofactors. We show that VCF1 engages in joint p97 complex formation with the heterodimeric primary p97 cofactor UFD1-NPL4 and promotes p97-UFD1-NPL4-dependent proteasomal degradation of ubiquitylated substrates in cells. Mechanistically, VCF1 indirectly stimulates UFD1-NPL4 interactions with ubiquitin conjugates via its binding to p97 but has no intrinsic affinity for ubiquitin. Collectively, our findings establish VCF1 as an unconventional p97 cofactor that promotes p97-dependent protein turnover by facilitating p97-UFD1-NPL4 recruitment to ubiquitylated targets.
Collapse
Affiliation(s)
- Ann Schirin Mirsanaye
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Saskia Hoffmann
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Melanie Weisser
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Andreas Mund
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Blanca Lopez Mendez
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Dimitris Typas
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Johannes van den Boom
- Molecular Biology I, Faculty of Biology, University of Duisburg-Essen, 45117, Essen, Germany
| | - Bente Benedict
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Ivo A Hendriks
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Michael Lund Nielsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Hemmo Meyer
- Molecular Biology I, Faculty of Biology, University of Duisburg-Essen, 45117, Essen, Germany
| | - Julien P Duxin
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Guillermo Montoya
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Niels Mailand
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, DK-2200, Copenhagen, Denmark.
- Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
30
|
Cao H, Zhou X, Xu B, Hu H, Guo J, Ma Y, Wang M, Li N, Jun Z. Advances in the study of protein folding and endoplasmic reticulum-associated degradation in mammal cells. J Zhejiang Univ Sci B 2024; 25:212-232. [PMID: 38453636 PMCID: PMC10918413 DOI: 10.1631/jzus.b2300403] [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: 06/11/2023] [Accepted: 08/03/2023] [Indexed: 03/09/2024]
Abstract
The endoplasmic reticulum is a key site for protein production and quality control. More than one-third of proteins are synthesized and folded into the correct three-dimensional conformation in the endoplasmic reticulum. However, during protein folding, unfolded and/or misfolded proteins are prone to occur, which may lead to endoplasmic reticulum stress. Organisms can monitor the quality of the proteins produced by endoplasmic reticulum quality control (ERQC) and endoplasmic reticulum-associated degradation (ERAD), which maintain endoplasmic reticulum protein homeostasis by degrading abnormally folded proteins. The underlying mechanisms of protein folding and ERAD in mammals have not yet been fully explored. Therefore, this paper reviews the process and function of protein folding and ERAD in mammalian cells, in order to help clinicians better understand the mechanism of ERAD and to provide a scientific reference for the treatment of diseases caused by abnormal ERAD.
Collapse
Affiliation(s)
- Hong Cao
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Xuchang Zhou
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Bowen Xu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Han Hu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Jianming Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Yuwei Ma
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Miao Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Nan Li
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China.
| | - Zou Jun
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
31
|
Mannar D, Ahmed S, Subramaniam S. AAA ATPase protein-protein interactions as therapeutic targets in cancer. Curr Opin Cell Biol 2024; 86:102291. [PMID: 38056141 DOI: 10.1016/j.ceb.2023.102291] [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: 06/30/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023]
Abstract
AAA ATPases are a conserved group of enzymes that couple ATP hydrolysis to diverse activities critical for cellular homeostasis by targeted protein-protein interactions. Some of these interactions are potential therapeutic targets because of their role in cancers which rely on increased AAA ATPase activities for maintenance of genomic stability. Two well-characterized members of this family are p97/VCP and RUVBL ATPases where there is a growing understanding of their structure and function, as well as an emerging landscape of selective inhibitors. Here we highlight recent progress in this field, with particular emphasis on structural advances enabled by cryo-electron microscopy (cryo-EM).
Collapse
Affiliation(s)
- Dhiraj Mannar
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sana Ahmed
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sriram Subramaniam
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Gandeeva Therapeutics, Inc., Burnaby, BC V5C 6N5, Canada.
| |
Collapse
|
32
|
Li H, Ji Z, Paulo JA, Gygi SP, Rapoport TA. Bidirectional substrate shuttling between the 26S proteasome and the Cdc48 ATPase promotes protein degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572403. [PMID: 38187576 PMCID: PMC10769200 DOI: 10.1101/2023.12.20.572403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Most eukaryotic proteins are degraded by the 26S proteasome after modification with a polyubiquitin chain. Substrates lacking unstructured segments cannot be degraded directly and require prior unfolding by the Cdc48 ATPase (p97 or VCP in mammals) in complex with its ubiquitin-binding partner Ufd1-Npl4 (UN). Here, we use purified yeast components to reconstitute Cdc48-dependent degradation of well-folded model substrates by the proteasome. We show that a minimal system consists of the 26S proteasome, the Cdc48-UN ATPase complex, the proteasome cofactor Rad23, and the Cdc48 cofactors Ubx5 and Shp1. Rad23 and Ubx5 stimulate polyubiquitin binding to the 26S proteasome and the Cdc48-UN complex, respectively, allowing these machines to compete for substrates before and after their unfolding. Shp1 stimulates protein unfolding by the Cdc48-UN complex, rather than substrate recruitment. In vivo experiments confirm that many proteins undergo bidirectional substrate shuttling between the 26S proteasome and Cdc48 ATPase before being degraded.
Collapse
|
33
|
Williams C, Dong KC, Arkinson C, Martin A. Preparation of site-specifically fluorophore-labeled polyubiquitin chains for FRET studies of Cdc48 substrate processing. STAR Protoc 2023; 4:102659. [PMID: 37889757 PMCID: PMC10630674 DOI: 10.1016/j.xpro.2023.102659] [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: 06/15/2023] [Revised: 08/24/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
A critical step in the removal of polyubiquitinated proteins from macromolecular complexes and membranes for subsequent proteasomal degradation is the unfolding of an ubiquitin moiety by the cofactor Ufd1/Npl4 (UN) and its insertion into the Cdc48 ATPase for mechanical translocation. Here, we present a stepwise protocol for the assembly and purification of Lys48-linked ubiquitin chains that are fluorophore labeled at specific ubiquitin moieties and allow monitoring polyubiquitin engagement by the Cdc48-UN complex in a FRET-based assay. For complete details on the use and execution of this protocol, please refer to Williams et al. (2023).1.
Collapse
Affiliation(s)
- Cameron Williams
- Biophysics Graduate Group, University of California, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Ken C Dong
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California at Berkeley, Berkeley CA 94720
| | - Connor Arkinson
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California at Berkeley, Berkeley CA 94720
| | - Andreas Martin
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California at Berkeley, Berkeley CA 94720.
| |
Collapse
|
34
|
Braxton JR, Altobelli CR, Tucker MR, Tse E, Thwin AC, Arkin MR, Southworth DR. The p97/VCP adaptor UBXD1 drives AAA+ remodeling and ring opening through multi-domain tethered interactions. Nat Struct Mol Biol 2023; 30:2009-2019. [PMID: 37945741 PMCID: PMC10716044 DOI: 10.1038/s41594-023-01126-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 09/14/2023] [Indexed: 11/12/2023]
Abstract
p97, also known as valosin-containing protein, is an essential cytosolic AAA+ (ATPases associated with diverse cellular activities) hexamer that unfolds substrate polypeptides to support protein homeostasis and macromolecular disassembly. Distinct sets of p97 adaptors guide cellular functions but their roles in direct control of the hexamer are unclear. The UBXD1 adaptor localizes with p97 in critical mitochondria and lysosome clearance pathways and contains multiple p97-interacting domains. Here we identify UBXD1 as a potent p97 ATPase inhibitor and report structures of intact human p97-UBXD1 complexes that reveal extensive UBXD1 contacts across p97 and an asymmetric remodeling of the hexamer. Conserved VIM, UBX and PUB domains tether adjacent protomers while a connecting strand forms an N-terminal domain lariat with a helix wedged at the interprotomer interface. An additional VIM-connecting helix binds along the second (D2) AAA+ domain. Together, these contacts split the hexamer into a ring-open conformation. Structures, mutagenesis and comparisons to other adaptors further reveal how adaptors containing conserved p97-remodeling motifs regulate p97 ATPase activity and structure.
Collapse
Affiliation(s)
- Julian R Braxton
- Graduate Program in Chemistry and Chemical Biology, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Chad R Altobelli
- Graduate Program in Chemistry and Chemical Biology, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California San Francisco, San Francisco, CA, USA
| | - Maxwell R Tucker
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Graduate Program in Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Eric Tse
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Aye C Thwin
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Michelle R Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California San Francisco, San Francisco, CA, USA.
| | - Daniel R Southworth
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
35
|
Mah-Som AY, Daw J, Huynh D, Wu M, Creekmore BC, Burns W, Skinner SA, Holla ØL, Smeland MF, Planes M, Uguen K, Redon S, Bierhals T, Scholz T, Denecke J, Mensah MA, Sczakiel HL, Tichy H, Verheyen S, Blatterer J, Schreiner E, Thies J, Lam C, Spaeth CG, Pena L, Ramsey K, Narayanan V, Seaver LH, Rodriguez D, Afenjar A, Burglen L, Lee EB, Chou TF, Weihl CC, Shinawi MS. An autosomal-dominant childhood-onset disorder associated with pathogenic variants in VCP. Am J Hum Genet 2023; 110:1959-1975. [PMID: 37883978 PMCID: PMC10645565 DOI: 10.1016/j.ajhg.2023.10.007] [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: 06/10/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Valosin-containing protein (VCP) is an AAA+ ATPase that plays critical roles in multiple ubiquitin-dependent cellular processes. Dominant pathogenic variants in VCP are associated with adult-onset multisystem proteinopathy (MSP), which manifests as myopathy, bone disease, dementia, and/or motor neuron disease. Through GeneMatcher, we identified 13 unrelated individuals who harbor heterozygous VCP variants (12 de novo and 1 inherited) associated with a childhood-onset disorder characterized by developmental delay, intellectual disability, hypotonia, and macrocephaly. Trio exome sequencing or a multigene panel identified nine missense variants, two in-frame deletions, one frameshift, and one splicing variant. We performed in vitro functional studies and in silico modeling to investigate the impact of these variants on protein function. In contrast to MSP variants, most missense variants had decreased ATPase activity, and one caused hyperactivation. Other variants were predicted to cause haploinsufficiency, suggesting a loss-of-function mechanism. This cohort expands the spectrum of VCP-related disease to include neurodevelopmental disease presenting in childhood.
Collapse
Affiliation(s)
- Annelise Y Mah-Som
- Genetics Training Program, Harvard Medical School and Brigham & Women's Hospital, Boston, MA 02115, USA; Division of Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jil Daw
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Diana Huynh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mengcheng Wu
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Benjamin C Creekmore
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | | - Øystein L Holla
- Department of Medical Genetics, Telemark Hospital, 3710 Skien, Norway
| | - Marie F Smeland
- Department of Pediatric Rehabilitation, University Hospital of North Norway and the Arctic, University of Norway, 9019 Tromsø, Norway
| | - Marc Planes
- Service de Génétique Médicale et Biologie de la Reproduction, and Centre de Référence Déficiences Intellectuelles, Service de Pédiatrie, CHU de Brest, 29200 Brest, France
| | - Kevin Uguen
- Service de Génétique Médicale et Biologie de la Reproduction, and Centre de Référence Déficiences Intellectuelles, Service de Pédiatrie, CHU de Brest, 29200 Brest, France; University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France
| | - Sylvia Redon
- Service de Génétique Médicale et Biologie de la Reproduction, and Centre de Référence Déficiences Intellectuelles, Service de Pédiatrie, CHU de Brest, 29200 Brest, France; University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Tasja Scholz
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Jonas Denecke
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Martin A Mensah
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; BIH Biomedical Innovation Academy, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany; RG Development and Disease, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Henrike L Sczakiel
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; BIH Biomedical Innovation Academy, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany; RG Development and Disease, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Heidelis Tichy
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, 8010 Graz, Austria
| | - Sarah Verheyen
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, 8010 Graz, Austria
| | - Jasmin Blatterer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, 8010 Graz, Austria
| | - Elisabeth Schreiner
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, 8010 Graz, Austria
| | - Jenny Thies
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Christina Lam
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Christine G Spaeth
- Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Loren Pena
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Laurie H Seaver
- Corewell Health Helen Devos Children's Hospital, Department of Pediatrics and Human Development, Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA
| | - Diana Rodriguez
- Departement of Pediatric Neurology & Reference Centre for Congenital Malformations and Diseases of the Cerebellum, AP-HP.Sorbonne Université - Hôpital d'Enfants Armand-Trousseau, 75012 Paris, France
| | - Alexandra Afenjar
- Cerebellar Malformations and Congenital Diseases Reference Center and Neurogenetics Lab, Department of Genetics, Armand Trousseau Hospital, AP-HP Sorbonne Université, 75006 Paris, France
| | - Lydie Burglen
- Cerebellar Malformations and Congenital Diseases Reference Center and Neurogenetics Lab, Department of Genetics, Armand Trousseau Hospital, AP-HP Sorbonne Université, 75006 Paris, France
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Tsui-Fen Chou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Conrad C Weihl
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Marwan S Shinawi
- Division of Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
36
|
Han SI, Nakakuki M, Nakagawa Y, Wang Y, Araki M, Yamamoto Y, Tokiwa H, Takeda H, Mizunoe Y, Motomura K, Ohno H, Kainoh K, Murayama Y, Aita Y, Takeuchi Y, Osaki Y, Miyamoto T, Sekiya M, Matsuzaka T, Yahagi N, Sone H, Daitoku H, Sato R, Kawano H, Shimano H. Rhomboid protease RHBDL4/RHBDD1 cleaves SREBP-1c at endoplasmic reticulum monitoring and regulating fatty acids. PNAS NEXUS 2023; 2:pgad351. [PMID: 37954160 PMCID: PMC10637267 DOI: 10.1093/pnasnexus/pgad351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/02/2023] [Indexed: 11/14/2023]
Abstract
The endoplasmic reticulum (ER)-embedded transcription factors, sterol regulatory element-binding proteins (SREBPs), master regulators of lipid biosynthesis, are transported to the Golgi for proteolytic activation to tune cellular cholesterol levels and regulate lipogenesis. However, mechanisms by which the cell responds to the levels of saturated or unsaturated fatty acids remain underexplored. Here, we show that RHBDL4/RHBDD1, a rhomboid family protease, directly cleaves SREBP-1c at the ER. The p97/VCP, AAA-ATPase complex then acts as an auxiliary segregase to extract the remaining ER-embedded fragment of SREBP-1c. Importantly, the enzymatic activity of RHBDL4 is enhanced by saturated fatty acids (SFAs) but inhibited by polyunsaturated fatty acids (PUFAs). Genetic deletion of RHBDL4 in mice fed on a Western diet enriched in SFAs and cholesterol prevented SREBP-1c from inducing genes for lipogenesis, particularly for synthesis and incorporation of PUFAs, and secretion of lipoproteins. The RHBDL4-SREBP-1c pathway reveals a regulatory system for monitoring fatty acid composition and maintaining cellular lipid homeostasis.
Collapse
Affiliation(s)
- Song-Iee Han
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masanori Nakakuki
- Pharmaceutical Research Center, Mochida Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8524, Japan
| | - Yoshimi Nakagawa
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Yunong Wang
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masaya Araki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Hiroaki Tokiwa
- Laboratory of Organic Chemistry, Gifu Pharmaceutical University, Daigaku-Nishi, Gifu 501-1196, Japan
| | - Hiroyuki Takeda
- Division of Proteo Drug Discovery Sciences, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Yuhei Mizunoe
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kaori Motomura
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiroshi Ohno
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kenta Kainoh
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Murayama
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuichi Aita
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshinori Takeuchi
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshinori Osaki
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takafumi Miyamoto
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Motohiro Sekiya
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Naoya Yahagi
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hirohito Sone
- Department of Internal Medicine, Faculty of Medicine, Niigata University, Niigata, Niigata 951-8510, Japan
| | - Hiroaki Daitoku
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Ryuichiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, Nutri-Life Science Laboratory, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroyuki Kawano
- Pharmaceutical Research Center, Mochida Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8524, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
- Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
37
|
Tsioras K, Smith KC, Edassery SL, Garjani M, Li Y, Williams C, McKenna ED, Guo W, Wilen AP, Hark TJ, Marklund SL, Ostrow LW, Gilthorpe JD, Ichida JK, Kalb RG, Savas JN, Kiskinis E. Analysis of proteome-wide degradation dynamics in ALS SOD1 iPSC-derived patient neurons reveals disrupted VCP homeostasis. Cell Rep 2023; 42:113160. [PMID: 37776851 PMCID: PMC10785776 DOI: 10.1016/j.celrep.2023.113160] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/18/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023] Open
Abstract
Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS) through gain-of-function effects, yet the mechanisms by which misfolded mutant SOD1 (mutSOD1) protein impairs human motor neurons (MNs) remain unclear. Here, we use induced-pluripotent-stem-cell-derived MNs coupled to metabolic stable isotope labeling and mass spectrometry to investigate proteome-wide degradation dynamics. We find several proteins, including the ALS-causal valosin-containing protein (VCP), which predominantly acts in proteasome degradation and autophagy, that degrade slower in mutSOD1 relative to isogenic control MNs. The interactome of VCP is altered in mutSOD1 MNs in vitro, while VCP selectively accumulates in the affected motor cortex of ALS-SOD1 patients. Overexpression of VCP rescues mutSOD1 toxicity in MNs in vitro and in a C. elegans model in vivo, in part due to its ability to modulate the degradation of insoluble mutSOD1. Our results demonstrate that VCP contributes to mutSOD1-dependent degeneration, link two distinct ALS-causal genes, and highlight selective protein degradation impairment in ALS pathophysiology.
Collapse
Affiliation(s)
- Konstantinos Tsioras
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kevin C Smith
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Seby L Edassery
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mehraveh Garjani
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yichen Li
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Chloe Williams
- Department of Integrative Medical Biology, Umeå University, 90187 Umeå, Sweden
| | - Elizabeth D McKenna
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Wenxuan Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Anika P Wilen
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Timothy J Hark
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Lyle W Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | | | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Robert G Kalb
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeffrey N Savas
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
38
|
Rao B, Wang Q, Yao D, Xia Y, Li W, Xie Y, Li S, Cao M, Shen Y, Qin A, Zhao J, Cao Y. The cryo-EM structure of the human ERAD retrotranslocation complex. SCIENCE ADVANCES 2023; 9:eadi5656. [PMID: 37831771 PMCID: PMC10575581 DOI: 10.1126/sciadv.adi5656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/12/2023] [Indexed: 10/15/2023]
Abstract
Endoplasmic reticulum-associated degradation (ERAD) maintains protein homeostasis by retrieving misfolded proteins from the endoplasmic reticulum (ER) lumen into the cytosol for degradation. The retrotranslocation of misfolded proteins across the ER membrane is an energy-consuming process, with the detailed transportation mechanism still needing clarification. We determined the cryo-EM structures of the hetero-decameric complex formed by the Derlin-1 tetramer and the p97 hexamer. It showed an intriguing asymmetric complex and a putative coordinated squeezing movement in Derlin-1 and p97 parts. With the conformational changes of p97 induced by its ATP hydrolysis activities, the Derlin-1 channel could be torn into a "U" shape with a large opening to the lipidic environment, thereby forming an entry for the substrates in the ER membrane. The EM analysis showed that p97 formed a functional protein complex with Derlin-1, revealing the coupling mechanism between the ERAD retrotranslocation and the ATP hydrolysis activities.
Collapse
Affiliation(s)
- Bing Rao
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qian Wang
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
| | - Deqiang Yao
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ying Xia
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
| | - Wenguo Li
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
| | - Yuming Xie
- College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shaobai Li
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
| | - Mi Cao
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
| | - Yafeng Shen
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Department of Orthopaedics, Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jie Zhao
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Department of Orthopaedics, Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu Cao
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 115 Jinzun Road, Shanghai 200125, China
| |
Collapse
|
39
|
Ziff OJ, Harley J, Wang Y, Neeves J, Tyzack G, Ibrahim F, Skehel M, Chakrabarti AM, Kelly G, Patani R. Nucleocytoplasmic mRNA redistribution accompanies RNA binding protein mislocalization in ALS motor neurons and is restored by VCP ATPase inhibition. Neuron 2023; 111:3011-3027.e7. [PMID: 37480846 DOI: 10.1016/j.neuron.2023.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 05/09/2023] [Accepted: 06/22/2023] [Indexed: 07/24/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by nucleocytoplasmic mislocalization of the RNA-binding protein (RBP) TDP-43. However, emerging evidence suggests more widespread mRNA and protein mislocalization. Here, we employed nucleocytoplasmic fractionation, RNA sequencing, and mass spectrometry to investigate the localization of mRNA and protein in induced pluripotent stem cell-derived motor neurons (iPSMNs) from ALS patients with TARDBP and VCP mutations. ALS mutant iPSMNs exhibited extensive nucleocytoplasmic mRNA redistribution, RBP mislocalization, and splicing alterations. Mislocalized proteins exhibited a greater affinity for redistributed transcripts, suggesting a link between RBP mislocalization and mRNA redistribution. Notably, treatment with ML240, a VCP ATPase inhibitor, partially restored mRNA and protein localization in ALS mutant iPSMNs. ML240 induced changes in the VCP interactome and lysosomal localization and reduced oxidative stress and DNA damage. These findings emphasize the link between RBP mislocalization and mRNA redistribution in ALS motor neurons and highlight the therapeutic potential of VCP inhibition.
Collapse
Affiliation(s)
- Oliver J Ziff
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK; National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, WC1N 3BG London, UK.
| | - Jasmine Harley
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK; Institute of Molecular and Cell Biology, A(∗)STAR Research Entities, Singapore 138673, Singapore
| | - Yiran Wang
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Jacob Neeves
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Giulia Tyzack
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Fairouz Ibrahim
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK
| | - Mark Skehel
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK
| | | | - Gavin Kelly
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Rickie Patani
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK; National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, WC1N 3BG London, UK.
| |
Collapse
|
40
|
Jones NH, Liu Q, Urnavicius L, Dahan NE, Vostal LE, Kapoor TM. Allosteric activation of VCP, a AAA unfoldase, by small molecule mimicry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560478. [PMID: 37873168 PMCID: PMC10592943 DOI: 10.1101/2023.10.02.560478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The loss of function of AAA (ATPases associated with diverse cellular activities) mechanoenzymes has been linked to diseases, and small molecules that activate these proteins can be powerful tools to probe mechanisms and test therapeutic hypotheses. Unlike chemical inhibitors that can bind a single conformational state to block enzyme activity, activator binding must be permissive to different conformational states needed for enzyme function. However, we do not know how AAA proteins can be activated by small molecules. Here, we focus on valosin-containing protein (VCP)/p97, a AAA unfoldase whose loss of function has been linked to protein aggregation-based disorders, to identify druggable sites for chemical activators. We identified VCP Activator 1 (VA1), a compound that dose-dependently stimulates VCP ATPase activity up to ∼3-fold. Our cryo-EM studies resulted in structures (∼2.9-3.5 Å-resolution) of VCP in apo and ADP-bound states, and revealed VA1 binding an allosteric pocket near the C-terminus in both states. Engineered mutations in the VA1 binding site confer resistance to VA1, and furthermore, modulate VCP activity to a similar level as VA1-mediated activation. The VA1 binding site can alternatively be occupied by a phenylalanine residue in the VCP C-terminal tail, a motif that is post-translationally modified and interacts with cofactors. Together, our findings uncover a druggable allosteric site and a mechanism of enzyme regulation that can be tuned through small molecule mimicry. Significance The loss of function of valosin-containing protein (VCP/p97), a mechanoenzyme from the AAA superfamily that hydrolyzes ATP and uses the released energy to extract or unfold substrate proteins, is linked to protein aggregation-based disorders. However, druggable allosteric sites to activate VCP, or any AAA mechanoenzyme, have not been identified. Here, we report cryo-EM structures of VCP in two states in complex with VA1, a compound we identified that dose-dependently stimulates VCP's ATP hydrolysis activity. The VA1 binding site can also be occupied by a phenylalanine residue in the VCP C-terminal tail, suggesting that VA1 acts through mimicry of this interaction. Our study reveals a druggable allosteric site and a mechanism of enzyme regulation.
Collapse
|
41
|
Körner M, Meyer SR, Marincola G, Kern MJ, Grimm C, Schuelein-Voelk C, Fischer U, Hofmann K, Buchberger A. The FAM104 proteins VCF1/2 promote the nuclear localization of p97/VCP. eLife 2023; 12:e92409. [PMID: 37713320 PMCID: PMC10541173 DOI: 10.7554/elife.92409] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 09/10/2023] [Indexed: 09/17/2023] Open
Abstract
The ATPase p97 (also known as VCP, Cdc48) has crucial functions in a variety of important cellular processes such as protein quality control, organellar homeostasis, and DNA damage repair, and its de-regulation is linked to neuromuscular diseases and cancer. p97 is tightly controlled by numerous regulatory cofactors, but the full range and function of the p97-cofactor network is unknown. Here, we identify the hitherto uncharacterized FAM104 proteins as a conserved family of p97 interactors. The two human family members VCP nuclear cofactor family member 1 and 2 (VCF1/2) bind p97 directly via a novel, alpha-helical motif and associate with p97-UFD1-NPL4 and p97-UBXN2B complexes in cells. VCF1/2 localize to the nucleus and promote the nuclear import of p97. Loss of VCF1/2 results in reduced nuclear p97 levels, slow growth, and hypersensitivity to chemical inhibition of p97 in the absence and presence of DNA damage, suggesting that FAM104 proteins are critical regulators of nuclear p97 functions.
Collapse
Affiliation(s)
- Maria Körner
- University of Würzburg, Biocenter, Chair of Biochemistry IWürzburgGermany
| | - Susanne R Meyer
- University of Würzburg, Biocenter, Chair of Biochemistry IWürzburgGermany
| | | | - Maximilian J Kern
- Department of Molecular Cell Biology, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Clemens Grimm
- University of Würzburg, Biocenter, Chair of Biochemistry IWürzburgGermany
| | | | - Utz Fischer
- University of Würzburg, Biocenter, Chair of Biochemistry IWürzburgGermany
| | - Kay Hofmann
- Institute of Genetics, University of CologneCologneGermany
| | | |
Collapse
|
42
|
Batra S, Vaquer-Alicea J, Manon VA, Kashmer OM, Lemoff A, Cairns NJ, White CL, Diamond MI. VCP increases or decreases tau seeding using specific cofactors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555637. [PMID: 37693404 PMCID: PMC10491269 DOI: 10.1101/2023.08.30.555637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Background Neurodegenerative tauopathies may progress based on seeding by pathological tau assemblies, whereby an aggregate is released from one cell, gains entry to an adjacent or connected cell, and serves as a specific template for its own replication in the cytoplasm. In vitro seeding reactions typically take days, yet seeding into the complex cytoplasmic milieu can happen within hours. A cellular machinery might regulate this process, but potential players are unknown. Methods We used proximity labeling to identify factors that control seed amplification. We fused split-APEX2 to the C-terminus of tau repeat domain (RD) to reconstitute peroxidase activity upon seeded intracellular tau aggregation. We identified valosin containing protein (VCP/p97) 5h after seeding. Mutations in VCP underlie two neurodegenerative diseases, multisystem proteinopathy and vacuolar tauopathy, but its mechanistic role is unclear. We utilized tau biosensors, a cellular model for tau aggregation, to study the effects of VCP on tau seeding. Results VCP knockdown reduced tau seeding. However, distinct chemical inhibitors of VCP and the proteasome had opposing effects on aggregation, but only when given <8h of seed exposure. ML-240 increased seeding efficiency ~40x, whereas NMS-873 decreased seeding efficiency by 50%, and MG132 increased seeding ~10x. We screened VCP co-factors in HEK293 biosensor cells by genetic knockout or knockdown. Reduction of ATXN3, NSFL1C, UBE4B, NGLY1, and OTUB1 decreased tau seeding, as did NPLOC4, which also uniquely increased soluble tau levels. Reduction of FAF2 and UBXN6 increased tau seeding. Conclusions VCP uses distinct cofactors to determine seed replication efficiency, consistent with a dedicated cytoplasmic processing complex that directs seeds towards dissolution vs. amplification.
Collapse
Affiliation(s)
- Sushobhna Batra
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jaime Vaquer-Alicea
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Victor A Manon
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Omar M Kashmer
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nigel J Cairns
- Department of Clinical and Biological Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Charles L White
- Department of Pathology, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Neurology, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
43
|
Blueggel M, Kroening A, Kracht M, van den Boom J, Dabisch M, Goehring A, Kaschani F, Kaiser M, Bayer P, Meyer H, Beuck C. The UBX domain in UBXD1 organizes ubiquitin binding at the C-terminus of the VCP/p97 AAA-ATPase. Nat Commun 2023; 14:3258. [PMID: 37277335 DOI: 10.1038/s41467-023-38604-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
The AAA+ ATPase p97/VCP together with different sets of substrate-delivery adapters and accessory cofactor proteins unfolds ubiquitinated substrates to facilitate degradation by the proteasome. The UBXD1 cofactor is connected to p97-associated multisystem proteinopathy but its biochemical function and structural organization on p97 has remained largely elusive. Using a combination of crosslinking mass spectrometry and biochemical assays, we identify an extended UBX (eUBX) module in UBXD1 related to a lariat in another cofactor, ASPL. Of note, the UBXD1-eUBX intramolecularly associates with the PUB domain in UBXD1 close to the substrate exit pore of p97. The UBXD1 PUB domain can also bind the proteasomal shuttling factor HR23b via its UBL domain. We further show that the eUBX domain has ubiquitin binding activity and that UBXD1 associates with an active p97-adapter complex during substrate unfolding. Our findings suggest that the UBXD1-eUBX module receives unfolded ubiquitinated substrates after they exit the p97 channel and before hand-over to the proteasome. The interplay of full-length UBXD1 and HR23b and their function in the context of an active p97:UBXD1 unfolding complex remains to be studied in future work.
Collapse
Affiliation(s)
- Mike Blueggel
- Structural and Medicinal Biochemistry, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Alexander Kroening
- Molecular Biology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Matthias Kracht
- Molecular Biology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | | | - Matthias Dabisch
- Structural and Medicinal Biochemistry, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Anna Goehring
- Structural and Medicinal Biochemistry, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Farnusch Kaschani
- Chemical Biology and ACE Analytical Core Facility Essen, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Markus Kaiser
- Chemical Biology and ACE Analytical Core Facility Essen, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Peter Bayer
- Structural and Medicinal Biochemistry, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Hemmo Meyer
- Molecular Biology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Christine Beuck
- Structural and Medicinal Biochemistry, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
44
|
Braxton JR, Altobelli CR, Tucker MR, Tse E, Thwin AC, Arkin MR, Southworth DR. The p97/VCP adapter UBXD1 drives AAA+ remodeling and ring opening through multi-domain tethered interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540864. [PMID: 37292947 PMCID: PMC10245715 DOI: 10.1101/2023.05.15.540864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
p97/VCP is an essential cytosolic AAA+ ATPase hexamer that extracts and unfolds substrate polypeptides during protein homeostasis and degradation. Distinct sets of p97 adapters guide cellular functions but their roles in direct control of the hexamer are unclear. The UBXD1 adapter localizes with p97 in critical mitochondria and lysosome clearance pathways and contains multiple p97-interacting domains. We identify UBXD1 as a potent p97 ATPase inhibitor and report structures of intact p97:UBXD1 complexes that reveal extensive UBXD1 contacts across p97 and an asymmetric remodeling of the hexamer. Conserved VIM, UBX, and PUB domains tether adjacent protomers while a connecting strand forms an N-terminal domain lariat with a helix wedged at the interprotomer interface. An additional VIM-connecting helix binds along the second AAA+ domain. Together these contacts split the hexamer into a ring-open conformation. Structures, mutagenesis, and comparisons to other adapters further reveal how adapters containing conserved p97-remodeling motifs regulate p97 ATPase activity and structure.
Collapse
Affiliation(s)
- Julian R. Braxton
- Graduate Program in Chemistry and Chemical Biology; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Chad R. Altobelli
- Graduate Program in Chemistry and Chemical Biology; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Maxwell R. Tucker
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
- Graduate Program in Biophysics; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Eric Tse
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Aye C. Thwin
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Michelle R. Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Daniel R. Southworth
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| |
Collapse
|
45
|
van den Berg SJW, East S, Mitra S, Jansen LET. p97/VCP drives turnover of SUMOylated centromeric CCAN proteins and CENP-A. Mol Biol Cell 2023; 34:br6. [PMID: 36989032 PMCID: PMC10162411 DOI: 10.1091/mbc.e23-01-0035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
The centromere is a unique chromatin domain that links sister chromatids and forms the attachment site for spindle microtubules in mitosis. Centromere inheritance is largely DNA sequence-independent but strongly reliant on a self-propagating chromatin domain featuring nucleosomes containing the H3 variant CENP-A. Unlike other histones, CENP-A is maintained with unusually high stability in chromatin. Previously, we have shown that mitotic maintenance of CENP-A and other constitutive centromere-associated network (CCAN) proteins is controlled by a dynamic SUMO cycle and that the deSUMOylase SENP6 is necessary for stable maintenance of CENP-A at the centromere. Here, we discover that the removal of SENP6 leads to a rapid loss of the CCAN, followed by a delayed loss of centromeric CENP-A, indicating that the CCAN is the primary SUMO target. We found that the ATP-dependent segregase p97/VCP removes centromeric CENP-A in a SUMO-dependent manner and interacts physically with the CCAN and CENP-A chromatin. Our data suggest a direct role of p97 in removing centromeric CENP-A via SUMOylated CCAN proteins, thereby ensuring centromere homeostasis and potentially preventing ectopic CENP-A accumulation.
Collapse
Affiliation(s)
- Sebastiaan J. W. van den Berg
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
- Instituto Gulbenkian de Ciencia, 2780-156 Oeiras, Portugal
| | - Samuel East
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Sreyoshi Mitra
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Lars E. T. Jansen
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| |
Collapse
|
46
|
Valimehr S, Sethi A, Shukla M, Bhattacharyya S, Kazemi M, Rouiller I. Molecular Mechanisms Driving and Regulating the AAA+ ATPase VCP/p97, an Important Therapeutic Target for Treating Cancer, Neurological and Infectious Diseases. Biomolecules 2023; 13:biom13050737. [PMID: 37238606 DOI: 10.3390/biom13050737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/15/2023] [Accepted: 04/13/2023] [Indexed: 05/28/2023] Open
Abstract
p97/VCP, a highly conserved type II ATPase associated with diverse cellular activities (AAA+ ATPase), is an important therapeutic target in the treatment of neurodegenerative diseases and cancer. p97 performs a variety of functions in the cell and facilitates virus replication. It is a mechanochemical enzyme that generates mechanical force from ATP-binding and hydrolysis to perform several functions, including unfolding of protein substrates. Several dozens of cofactors/adaptors interact with p97 and define the multifunctionality of p97. This review presents the current understanding of the molecular mechanism of p97 during the ATPase cycle and its regulation by cofactors and small-molecule inhibitors. We compare detailed structural information obtained in different nucleotide states in the presence and absence of substrates and inhibitors. We also review how pathogenic gain-of-function mutations modify the conformational changes of p97 during the ATPase cycle. Overall, the review highlights how the mechanistic knowledge of p97 helps in designing pathway-specific modulators and inhibitors.
Collapse
Affiliation(s)
- Sepideh Valimehr
- Department of Biochemistry & Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- Bio21 Ian Holmes Imaging Centre, Department of Biochemistry & Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Ashish Sethi
- Department of Biochemistry & Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- Australian Nuclear Science Technology Organisation, The Australian Synchrotron, 800 Blackburn Rd, Clayton, VIC 3168, Australia
| | - Manjari Shukla
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur 342030, Rajasthan, India
| | - Sudipta Bhattacharyya
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur 342030, Rajasthan, India
| | - Mohsen Kazemi
- Department of Biochemistry & Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Isabelle Rouiller
- Department of Biochemistry & Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
47
|
Scalia P, Merali C, Barrero C, Suma A, Carnevale V, Merali S, Williams SJ. Novel Isoform DTX3c Associates with UBE2N-UBA1 and Cdc48/p97 as Part of the EphB4 Degradation Complex Regulated by the Autocrine IGF-II/IR A Signal in Malignant Mesothelioma. Int J Mol Sci 2023; 24:ijms24087380. [PMID: 37108544 PMCID: PMC10139083 DOI: 10.3390/ijms24087380] [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: 02/22/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
EphB4 angiogenic kinase over-expression in Mesothelioma cells relies upon a degradation rescue signal provided by autocrine IGF-II activation of Insulin Receptor A. However, the identity of the molecular machinery involved in EphB4 rapid degradation upon IGF-II signal deprivation are unknown. Using targeted proteomics, protein-protein interaction methods, PCR cloning, and 3D modeling approaches, we identified a novel ubiquitin E3 ligase complex recruited by the EphB4 C tail upon autocrine IGF-II signal deprivation. We show this complex to contain a previously unknown N-Terminal isoform of Deltex3 E3-Ub ligase (referred as "DTX3c"), along with UBA1(E1) and UBE2N(E2) ubiquitin ligases and the ATPase/unfoldase Cdc48/p97. Upon autocrine IGF-II neutralization in cultured MSTO211H (a Malignant Mesothelioma cell line that is highly responsive to the EphB4 degradation rescue IGF-II signal), the inter-molecular interactions between these factors were enhanced and their association with the EphB4 C-tail increased consistently with the previously described EphB4 degradation pattern. The ATPase/unfoldase activity of Cdc48/p97 was required for EphB4 recruitment. As compared to the previously known isoforms DTX3a and DTX3b, a 3D modeling analysis of the DTX3c Nt domain showed a unique 3D folding supporting isoform-specific biological function(s). We shed light on the molecular machinery associated with autocrine IGF-II regulation of oncogenic EphB4 kinase expression in a previously characterized IGF-II+/EphB4+ Mesothelioma cell line. The study provides early evidence for DTX3 Ub-E3 ligase involvement beyond the Notch signaling pathway.
Collapse
Affiliation(s)
- Pierluigi Scalia
- The ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA 19102, USA and 93100 Caltanissetta, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Carmen Merali
- Proteomics and Metabolomics Facility, Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| | - Carlos Barrero
- Proteomics and Metabolomics Facility, Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| | - Antonio Suma
- Institute of Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Vincenzo Carnevale
- Institute of Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Salim Merali
- Proteomics and Metabolomics Facility, Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| | - Stephen J Williams
- The ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA 19102, USA and 93100 Caltanissetta, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
48
|
Phan JM, Creekmore BC, Nguyen AT, Bershadskaya DD, Darwich NF, Lee EB. Novel VCP activator reverses multisystem proteinopathy nuclear proteostasis defects and enhances TDP-43 aggregate clearance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532082. [PMID: 36993559 PMCID: PMC10055171 DOI: 10.1101/2023.03.15.532082] [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: 04/29/2023]
Abstract
Pathogenic variants in VCP cause multisystem proteinopathy (MSP), a disease characterized by multiple clinical phenotypes including inclusion body myopathy, Paget's disease of the bone, and frontotemporal dementia (FTD). How such diverse phenotypes are driven by pathogenic VCP variants is not known. We found that these diseases exhibit a common pathologic feature, ubiquitinated intranuclear inclusions affecting myocytes, osteoclasts and neurons. Moreover, knock-in cell lines harboring MSP variants show a reduction in nuclear VCP. Given that MSP is associated with neuronal intranuclear inclusions comprised of TDP-43 protein, we developed a cellular model whereby proteostatic stress results in the formation of insoluble intranuclear TDP-43 aggregates. Consistent with a loss of nuclear VCP function, cells harboring MSP variants or cells treated with VCP inhibitor exhibited decreased clearance of insoluble intranuclear TDP-43 aggregates. Moreover, we identified four novel compounds that activate VCP primarily by increasing D2 ATPase activity whereby pharmacologic VCP activation appears to enhance clearance of insoluble intranuclear TDP-43 aggregate. Our findings suggest that VCP function is important for nuclear protein homeostasis, that MSP may be the result of impaired nuclear proteostasis, and that VCP activation may be potential therapeutic by virtue of enhancing the clearance of intranuclear protein aggregates.
Collapse
Affiliation(s)
- Jessica M Phan
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, PA, USA
| | - Benjamin C Creekmore
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, PA, USA
| | - Aivi T Nguyen
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, PA, USA
| | - Darya D Bershadskaya
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, PA, USA
| | - Nabil F Darwich
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, PA, USA
| | - Edward B Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, PA, USA
| |
Collapse
|
49
|
Williams C, Dong KC, Arkinson C, Martin A. The Ufd1 cofactor determines the linkage specificity of polyubiquitin chain engagement by the AAA+ ATPase Cdc48. Mol Cell 2023; 83:759-769.e7. [PMID: 36736315 PMCID: PMC9992269 DOI: 10.1016/j.molcel.2023.01.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/13/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023]
Abstract
The AAA+ ATPase Cdc48 utilizes the cofactor Ufd1/Npl4 to bind and thread polyubiquitinated substrates for their extraction from complexes or membranes and often for subsequent proteasomal degradation. Previous studies indicated that Cdc48 engages polyubiquitin chains through the Npl4-mediated unfolding of an initiator ubiquitin; yet, the underlying principles remain largely unknown. Using FRET-based assays, we revealed the mechanisms and kinetics of ubiquitin unfolding, insertion into the ATPase, and unfolding of the ubiquitin-attached substrate. We found that Cdc48 uses Ufd1's UT3 domain to bind a K48-linked ubiquitin on the initiator's proximal side of the chain, thereby directing the initiator toward rapid unfolding by Npl4 and engagement by Cdc48. Ubiquitins on the initiator's distal side increase substrate affinity and facilitate unfolding but impede substrate release from Cdc48-Ufd1/Npl4 in the absence of additional cofactors. Our findings explain how Cdc48-UN efficiently processes substrates with K48-linked chains of 4-6 ubiquitins, which represent most cellular polyubiquitinated proteins.
Collapse
Affiliation(s)
- Cameron Williams
- Biophysics Graduate Group, University of California at Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Ken C Dong
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Connor Arkinson
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Andreas Martin
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
50
|
Meyer H, van den Boom J. Targeting of client proteins to the VCP/p97/Cdc48 unfolding machine. Front Mol Biosci 2023; 10:1142989. [PMID: 36825201 PMCID: PMC9941556 DOI: 10.3389/fmolb.2023.1142989] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
The AAA+ ATPase p97 (also called VCP or Cdc48) is a major protein unfolding machine with hundreds of clients in diverse cellular pathways that are critical for cell homeostasis, proliferation and signaling. In this review, we summarize recent advances in understanding how diverse client proteins are targeted to the p97 machine to facilitate client degradation or to strip clients from binding partners for regulation. We describe an elaborate system that is governed by at least two types of alternative adapters. The Ufd1-Npl4 adapter along with accessory adapters targets ubiquitylated clients in the majority of pathways and uses ubiquitin as a universal unfolding tag. In contrast, the family of SEP-domain adapters such as p37 can target clients directly to p97 in a ubiquitin-independent manner. Despite the different targeting strategies, both pathways converge by inserting the client into the p97 pore to initiate a peptide threading mechanism through the central channel of p97 that drives client protein unfolding, protein extraction from membranes and protein complex disassembly processes.
Collapse
Affiliation(s)
- Hemmo Meyer
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | | |
Collapse
|