1
|
Liu HN, Wang T, Hu JJ, Chen L, Shi X, Li YM, Luo SZ. The disordered protein SERF promotes α-Synuclein aggregation through liquid-liquid phase separation. J Biol Chem 2024; 300:105667. [PMID: 38272228 PMCID: PMC10877630 DOI: 10.1016/j.jbc.2024.105667] [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: 03/01/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
The aggregation of α-Synuclein (α-Syn) into amyloid fibrils is the hallmark of Parkinson's disease. Under stress or other pathological conditions, the accumulation of α-Syn oligomers is the main contributor to the cytotoxicity. A potential approach for treating Parkinson's disease involves preventing the accumulation of these α-Syn oligomers. In this study, we present a novel mechanism involving a conserved group of disorderly proteins known as small EDRK-rich factor (SERF), which promotes the aggregation of α-Syn through a cophase separation process. Using diverse methods like confocal microscopy, fluorescence recovery after photobleaching assays, solution-state NMR spectroscopy, and Western blot, we determined that the N-terminal domain of SERF1a plays a role in the interactions that occur during cophase separation. Within these droplets, α-Syn undergoes a gradual transformation from solid condensates to amyloid fibrils, while SERF1a is excluded from the condensates and dissolves into the solution. Notably, in vivo experiments show that SERF1a cophase separation with α-Syn significantly reduces the deposition of α-Syn oligomers and decreases its cellular toxicity under stress. These findings suggest that SERF1a accelerates the conversion of α-Syn from highly toxic oligomers to less toxic fibrils through cophase separation, thereby mitigating the biological damage of α-Syn aggregation.
Collapse
Affiliation(s)
- He-Ning Liu
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ting Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jin-Jian Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Long Chen
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xiangyan Shi
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, China.
| | - Yan-Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China.
| | - Shi-Zhong Luo
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
2
|
Wang J, Dai L, Chen S, Zhang Z, Fang X, Zhang Z. Protein-protein interactions regulating α-synuclein pathology. Trends Neurosci 2024; 47:209-226. [PMID: 38355325 DOI: 10.1016/j.tins.2024.01.002] [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: 09/11/2023] [Revised: 12/15/2023] [Accepted: 01/21/2024] [Indexed: 02/16/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the formation of Lewy bodies (LBs). The main proteinaceous component of LBs is aggregated α-synuclein (α-syn). However, the mechanisms underlying α-syn aggregation are not yet fully understood. Converging lines of evidence indicate that, under certain pathological conditions, various proteins can interact with α-syn and regulate its aggregation. Understanding these protein-protein interactions is crucial for unraveling the molecular mechanisms contributing to PD pathogenesis. In this review we provide an overview of the current knowledge on protein-protein interactions that regulate α-syn aggregation. Additionally, we briefly summarize the methods used to investigate the influence of protein-protein interactions on α-syn aggregation and propagation.
Collapse
Affiliation(s)
- Jiannan Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sichun Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xin Fang
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China.
| |
Collapse
|
3
|
Qi S, Peng Y, Wang G, Zhang X, Liu M, He L. A tale of dual functions of SERF family proteins in regulating amyloid formation. Chembiochem 2024; 25:e202300727. [PMID: 38100267 DOI: 10.1002/cbic.202300727] [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: 10/23/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/17/2023]
Abstract
The abnormal aggregation of proteins is a significant pathological hallmark of diseases, such as the amyloid formation associated with fused in sarcoma protein (FUS) in frontotemporal lobar degeneration and amyotrophic lateral sclerosis diseases. Understanding which cellular components and how these components regulate the process of abnormal protein aggregation in living organisms is crucial for the prevention and treatment of neurodegenerative diseases. MOAG-4/SERF is a conserved family of proteins with rich positive charged residues, which was initially identified as an enhancer for the formation of amyloids in C. elegans. Knocking out SERF impedes the amyloid formation of various proteins, including α-synuclein and β-amyloid, which are linked to Parkinson's and Alzheimer's diseases, respectively. However, recent studies revealed SERF exhibited dual functions, as it could both promote and inhibit the fibril formation of the neurodegenerative disease-related amyloidogenic proteins. The connection between functions and structure basis of SERF in regulating the amyloid formation is still unclear. This review will outline the hallmark proteins in neurodegenerative diseases, summarize the contradictory role of the SERF protein family in promoting and inhibiting the aggregation of neurodegenerative proteins, and finally explore the potential structural basis and functional selectivity of the SERF protein.
Collapse
Affiliation(s)
- Shixing Qi
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yun Peng
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Guan Wang
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xu Zhang
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Optics Valley Laboratory, Wu Han Shi, 430074, Hubei, China
| | - Lichun He
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| |
Collapse
|
4
|
Tsai TY, Jhang WT, Hsu HK, Chan YT, Chang CF, Chen YR. Amyloid Modifier SERF1a Accelerates Alzheimer's Amyloid-β Fibrillization and Exacerbates the Cytotoxicity. ACS Chem Neurosci 2024; 15:479-490. [PMID: 38211979 DOI: 10.1021/acschemneuro.3c00403] [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] [Indexed: 01/13/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating, progressive neurodegenerative disease affecting the elderly in the world. The pathological hallmark senile plaques are mainly composed of amyloid-β (Aβ), in which the main isoforms are Aβ40 and Aβ42. Aβ is prone to aggregate and ultimately forms amyloid fibrils in the brains of AD patients. Factors that alter the Aβ aggregation process have been considered to be potential targets for treatments of AD. Modifier of aggregation 4 (MOAG-4)/small EDRK-rich factor (SERF) was previously selected from a chemical mutagenesis screen and identified as an amyloid modifier that promotes amyloid aggregation for α-synuclein, huntingtin, and Aβ40. The interaction and effect of yeast ScSERF on Aβ40 were previously described. Here, we examined the human SERF1a effect on Aβ40 and Aβ42 fibrillization by the Thioflavin T assay and found that SERF1a accelerated Aβ fibrillization in a dose-dependent manner without changing the fibril amount and without incorporation. By Fourier transform infrared spectroscopy (FTIR) and transmission electron microscopy (TEM), we found that SERF1a altered the secondary structures and the morphology of Aβ fibrils. The electrospray ionization mass spectrometry (ESI-MS) and analytical ultracentrifugation (AUC) results showed that SERF1a binds to Aβ in a 1:1 stoichiometry. Moreover, the NMR study showed that SERF1a interacts with Aβ via its N-terminal region. Cytotoxicity assay demonstrated that SERF1a enhanced toxicity of Aβ intermediates, and the effect can be rescued by SERF1a antibody. Overall, our study provides the underlying molecular mechanism for the SERF1a effect on Aβ fibrillization and facilitates the therapeutic development of AD.
Collapse
Affiliation(s)
- Tien-Ying Tsai
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 115, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Biological Chemistry, Academia Sinica, 128, Academia Road, Sec. 2, Nankang District, Taipei 115, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Wei-Ting Jhang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Hung-Kai Hsu
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yi-Tsu Chan
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Chi-Fon Chang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 115, Taiwan
| | - Yun-Ru Chen
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 115, Taiwan
| |
Collapse
|
5
|
Lurette O, Martín-Jiménez R, Khan M, Sheta R, Jean S, Schofield M, Teixeira M, Rodriguez-Aller R, Perron I, Oueslati A, Hebert-Chatelain E. Aggregation of alpha-synuclein disrupts mitochondrial metabolism and induce mitophagy via cardiolipin externalization. Cell Death Dis 2023; 14:729. [PMID: 37949858 PMCID: PMC10638290 DOI: 10.1038/s41419-023-06251-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Accumulation of α-synuclein aggregates in the substantia nigra pars compacta is central in the pathophysiology of Parkinson's disease, leading to the degeneration of dopaminergic neurons and the manifestation of motor symptoms. Although several PD models mimic the pathological accumulation of α-synuclein after overexpression, they do not allow for controlling and monitoring its aggregation. We recently generated a new optogenetic tool by which we can spatiotemporally control the aggregation of α-synuclein using a light-induced protein aggregation system. Using this innovative tool, we aimed to characterize the impact of α-synuclein clustering on mitochondria, whose activity is crucial to maintain neuronal survival. We observed that aggregates of α-synuclein transiently and dynamically interact with mitochondria, leading to mitochondrial depolarization, lower ATP production, mitochondrial fragmentation and degradation via cardiolipin externalization-dependent mitophagy. Aggregation of α-synuclein also leads to lower mitochondrial content in human dopaminergic neurons and in mouse midbrain. Interestingly, overexpression of α-synuclein alone did not induce mitochondrial degradation. This work is among the first to clearly discriminate between the impact of α-synuclein overexpression and aggregation on mitochondria. This study thus represents a new framework to characterize the role of mitochondria in PD.
Collapse
Affiliation(s)
- Olivier Lurette
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Rebeca Martín-Jiménez
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Mehtab Khan
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Razan Sheta
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Stéphanie Jean
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Mia Schofield
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Maxime Teixeira
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Raquel Rodriguez-Aller
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Isabelle Perron
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Abid Oueslati
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Etienne Hebert-Chatelain
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada.
- Department of Biology, University of Moncton, Moncton, NB, Canada.
| |
Collapse
|
6
|
Merle DA, Sen M, Armento A, Stanton CM, Thee EF, Meester-Smoor MA, Kaiser M, Clark SJ, Klaver CCW, Keane PA, Wright AF, Ehrmann M, Ueffing M. 10q26 - The enigma in age-related macular degeneration. Prog Retin Eye Res 2023; 96:101154. [PMID: 36513584 DOI: 10.1016/j.preteyeres.2022.101154] [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: 09/14/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
Despite comprehensive research efforts over the last decades, the pathomechanisms of age-related macular degeneration (AMD) remain far from being understood. Large-scale genome wide association studies (GWAS) were able to provide a defined set of genetic aberrations which contribute to disease risk, with the strongest contributors mapping to distinct regions on chromosome 1 and 10. While the chromosome 1 locus comprises factors of the complement system with well-known functions, the role of the 10q26-locus in AMD-pathophysiology remains enigmatic. 10q26 harbors a cluster of three functional genes, namely PLEKHA1, ARMS2 and HTRA1, with most of the AMD-associated genetic variants mapping to the latter two genes. High linkage disequilibrium between ARMS2 and HTRA1 has kept association studies from reliably defining the risk-causing gene for long and only very recently the genetic risk region has been narrowed to ARMS2, suggesting that this is the true AMD gene at this locus. However, genetic associations alone do not suffice to prove causality and one or more of the 14 SNPs on this haplotype may be involved in long-range control of gene expression, leaving HTRA1 and PLEKHA1 still suspects in the pathogenic pathway. Both, ARMS2 and HTRA1 have been linked to extracellular matrix homeostasis, yet their exact molecular function as well as their role in AMD pathogenesis remains to be uncovered. The transcriptional regulation of the 10q26 locus adds an additional level of complexity, given, that gene-regulatory as well as epigenetic alterations may influence expression levels from 10q26 in diseased individuals. Here, we provide a comprehensive overview on the 10q26 locus and its three gene products on various levels of biological complexity and discuss current and future research strategies to shed light on one of the remaining enigmatic spots in the AMD landscape.
Collapse
Affiliation(s)
- David A Merle
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department of Ophthalmology, Medical University of Graz, 8036, Graz, Austria.
| | - Merve Sen
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany
| | - Angela Armento
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany
| | - Chloe M Stanton
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Eric F Thee
- Department of Ophthalmology, Erasmus University Medical Center, 3015GD, Rotterdam, Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015CE, Rotterdam, Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus University Medical Center, 3015GD, Rotterdam, Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015CE, Rotterdam, Netherlands
| | - Markus Kaiser
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, 45117, Essen, Germany
| | - Simon J Clark
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus University Medical Center, 3015GD, Rotterdam, Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015CE, Rotterdam, Netherlands; Department of Ophthalmology, Radboudumc, 6525EX, Nijmegen, Netherlands; Institute of Molecular and Clinical Ophthalmology Basel, CH-4031, Basel, Switzerland
| | - Pearse A Keane
- Institute for Health Research, Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital NHS Foundation Trust, UCL Institute of Ophthalmology, London, EC1V 2PD, UK
| | - Alan F Wright
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Michael Ehrmann
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, 45117, Essen, Germany
| | - Marius Ueffing
- Institute for Ophthalmic Research, Department for Ophthalmology, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
7
|
Sahoo BR, Bardwell JCA. SERF, a family of tiny highly conserved, highly charged proteins with enigmatic functions. FEBS J 2023; 290:4150-4162. [PMID: 35694898 DOI: 10.1111/febs.16555] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/27/2022]
Abstract
Amyloid formation is a misfolding process that has been linked to age-related diseases, including Alzheimer's and Huntington's. Understanding how cellular factors affect this process in vivo is vital in realizing the dream of controlling this insidious process that robs so many people of their humanity. SERF (small EDRK-rich factor) was initially isolated as a factor that accelerated polyglutamine amyloid formation in a C. elegans model. SERF knockouts inhibit amyloid formation of a number of proteins that include huntingtin, α-synuclein and β-amyloid which are associated with Huntington's, Parkinson's and Alzheimer's disease, respectively, and purified SERF protein speeds their amyloid formation in vitro. SERF proteins are highly conserved, highly charged and conformationally dynamic proteins that form a fuzzy complex with amyloid precursors. They appear to act by specifically accelerating the primary step of amyloid nucleation. Brain-specific SERF knockout mice, though viable, appear to be more prone to deposition of amyloids, and show modified fibril morphology. Whole-body knockouts are perinatally lethal due to an apparently unrelated developmental issue. Recently, it was found that SERF binds RNA and is localized to nucleic acid-rich membraneless compartments. SERF-related sequences are commonly found fused to zinc finger sequences. These results point towards a nucleic acid-binding function. How this function relates to their ability to accelerate amyloid formation is currently obscure. In this review, we discuss the possible biological functions of SERF family proteins in the context of their structural fuzziness, modulation of amyloid pathway, nucleic acid binding and their fusion to folded proteins.
Collapse
Affiliation(s)
- Bikash R Sahoo
- Department of Molecular, Cellular and Developmental Biology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - James C A Bardwell
- Department of Molecular, Cellular and Developmental Biology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Tsai TY, Chen CY, Lin TW, Lin TC, Chiu FL, Shih O, Chang MY, Lin YC, Su AC, Chen CM, Jeng US, Kuo HC, Chang CF, Chen YR. Amyloid modifier SERF1a interacts with polyQ-expanded huntingtin-exon 1 via helical interactions and exacerbates polyQ-induced toxicity. Commun Biol 2023; 6:767. [PMID: 37479809 PMCID: PMC10361993 DOI: 10.1038/s42003-023-05142-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 07/13/2023] [Indexed: 07/23/2023] Open
Abstract
Abnormal polyglutamine (polyQ) expansion and fibrillization occur in Huntington's disease (HD). Amyloid modifier SERF enhances amyloid formation, but the underlying mechanism is not revealed. Here, the fibrillization and toxicity effect of SERF1a on Htt-exon1 are examined. SERF1a enhances the fibrillization of and interacts with mutant thioredoxin (Trx)-fused Httex1. NMR studies with Htt peptides show that TrxHttex1-39Q interacts with the helical regions in SERF1a and SERF1a preferentially interacts with the N-terminal 17 residues of Htt. Time-course analysis shows that SERF1a induces mutant TrxHttex1 to a single conformation enriched of β-sheet. Co-expression of SERF1a and Httex1-polyQ in neuroblastoma and lentiviral infection of SERF1a in HD-induced polypotent stem cell (iPSC)-derived neurons demonstrates the detrimental effect of SERF1a in HD. Higher level of SERF1a transcript or protein is detected in HD iPSC, transgenic mice, and HD plasma. Overall, this study provides molecular mechanism for SERF1a and mutant Httex1 to facilitate therapeutic development for HD.
Collapse
Affiliation(s)
- Tien-Ying Tsai
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Biological Chemistry, Academia Sinica, 128, Academia Road, Sec. 2. Nankang, Taipei, 115, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Chun-Yu Chen
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - Tien-Wei Lin
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - Tien-Chang Lin
- National Synchrotron Radiation Research Center, Hsinchu, 300, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Feng-Lan Chiu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - Orion Shih
- National Synchrotron Radiation Research Center, Hsinchu, 300, Taiwan
| | - Ming-Yun Chang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yu-Chun Lin
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - An-Chung Su
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - U-Ser Jeng
- National Synchrotron Radiation Research Center, Hsinchu, 300, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - Chi-Fon Chang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - Yun-Ru Chen
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei, 115, Taiwan.
| |
Collapse
|
9
|
Stroo E, Janssen L, Sin O, Hogewerf W, Koster M, Harkema L, Youssef SA, Beschorner N, Wolters AH, Bakker B, Becker L, Garrett L, Marschall S, Hoelter SM, Wurst W, Fuchs H, Gailus-Durner V, Hrabe de Angelis M, Thathiah A, Foijer F, van de Sluis B, van Deursen J, Jucker M, de Bruin A, Nollen EA. Deletion of SERF2 in mice delays embryonic development and alters amyloid deposit structure in the brain. Life Sci Alliance 2023; 6:e202201730. [PMID: 37130781 PMCID: PMC10155860 DOI: 10.26508/lsa.202201730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/04/2023] Open
Abstract
In age-related neurodegenerative diseases, like Alzheimer's and Parkinson's, disease-specific proteins become aggregation-prone and form amyloid-like deposits. Depletion of SERF proteins ameliorates this toxic process in worm and human cell models for diseases. Whether SERF modifies amyloid pathology in mammalian brain, however, has remained unknown. Here, we generated conditional Serf2 knockout mice and found that full-body deletion of Serf2 delayed embryonic development, causing premature birth and perinatal lethality. Brain-specific Serf2 knockout mice, on the other hand, were viable, and showed no major behavioral or cognitive abnormalities. In a mouse model for amyloid-β aggregation, brain depletion of Serf2 altered the binding of structure-specific amyloid dyes, previously used to distinguish amyloid polymorphisms in the human brain. These results suggest that Serf2 depletion changed the structure of amyloid deposits, which was further supported by scanning transmission electron microscopy, but further study will be required to confirm this observation. Altogether, our data reveal the pleiotropic functions of SERF2 in embryonic development and in the brain and support the existence of modifying factors of amyloid deposition in mammalian brain, which offer possibilities for polymorphism-based interventions.
Collapse
Affiliation(s)
- Esther Stroo
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Leen Janssen
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Olga Sin
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Graduate Program in Areas of Basic and Applied Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Wytse Hogewerf
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Mirjam Koster
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Liesbeth Harkema
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sameh A Youssef
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Natalie Beschorner
- Department of Cellular Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Anouk Hg Wolters
- Department of Biomedical Sciences of Cells and Systems, University Medical Centre Groningen, Groningen, The Netherlands
| | - Bjorn Bakker
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Lilian Garrett
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Susan Marschall
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Sabine M Hoelter
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Technische Universität München, Freising-Weihenstephan, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Developmental Genetics, TUM School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- Deutsches Institut für Neurodegenerative Erkrankungen (DZNE) Site Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Amantha Thathiah
- VIB Center for the Biology of Disease, KU Leuven Center for Human Genetics, University of Leuven, Leuven, Belgium
- Department of Neurobiology, University of Pittsburgh Brain Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | - Matthias Jucker
- Department of Cellular Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Alain de Bruin
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Ellen Aa Nollen
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
10
|
Belykh AE, Soldatov VO, Stetskaya TA, Kobzeva KA, Soldatova MO, Polonikov AV, Deykin AV, Churnosov MI, Freidin MB, Bushueva OY. Polymorphism of SERF2, the gene encoding a heat-resistant obscure (Hero) protein with chaperone activity, is a novel link in ischemic stroke. IBRO Neurosci Rep 2023; 14:453-461. [PMID: 37252629 PMCID: PMC10209486 DOI: 10.1016/j.ibneur.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/09/2023] [Indexed: 05/31/2023] Open
Abstract
Background Ischemic stroke (IS) is one of the most serious cardiovascular events associated with high risk of death or disability. The growing body of evidence highlights molecular chaperones as especially important players in the pathogenesis of the disease. Since six small proteins called "Hero" have been recently identified as a novel class of chaperones we aimed to evaluate whether SNP rs4644832 in SERF2 gene encoding the member of Hero-proteins, is associated with the risk of IS. Methods A total of 1929 unrelated Russians (861 patients with IS and 1068 healthy individuals) from Central Russia were recruited into the study. Genotyping was done using a probe-based PCR approach. Statistical analysis was carried out in the whole group and stratified by age, gender and smoking status. Results Analysis of the link between rs4644832 SERF2 and IS showed that G allele is the risk factor of IS only in females (OR=1.29, 95%CI 1.02-1.64, Padj=0.035). In addition, the analysis of associations of rs4644832 SERF2 and IS depending on the smoking status revealed that this genetic variant is associated with an increased risk of IS exclusively in non-smoking individuals (OR=1.26, 95%CI 1.01-1.56, P = 0.041). Discussion Sex- and smoking interactions between rs4644832 polymorphism and IS may be related to the impact of tobacco components metabolism and sex hormones on SERF2 expression. Conclusion The present study reveals the novel genetic association between rs4644832 polymorphism and the risk of IS suggesting that SERF2, the part of the protein quality control system, contributes to the pathogenesis of the disease.
Collapse
Affiliation(s)
- Andrei E. Belykh
- Pathophysiology Department, Kursk State Medical University, Kursk, Russia
| | - Vladislav O. Soldatov
- Laboratory of Genome Editing for Veterinary and Biomedicine, Belgorod State National Research University, Belgorod, Russia
| | - Tatiana A. Stetskaya
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
| | - Ksenia A. Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
| | - Maria O. Soldatova
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
| | - Alexey V. Polonikov
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russia
| | - Alexey V. Deykin
- Laboratory of Genome Editing for Veterinary and Biomedicine, Belgorod State National Research University, Belgorod, Russia
| | - Mikhail I. Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, Belgorod, Russia
| | - Maxim B. Freidin
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia
- Queen Mary University of London, London, United Kingdom
| | - Olga Y. Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russia
| |
Collapse
|
11
|
Lenard AJ, Mulder FAA, Madl T. Solvent paramagnetic relaxation enhancement as a versatile method for studying structure and dynamics of biomolecular systems. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2022; 132-133:113-139. [PMID: 36496256 DOI: 10.1016/j.pnmrs.2022.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/17/2023]
Abstract
Solvent paramagnetic relaxation enhancement (sPRE) is a versatile nuclear magnetic resonance (NMR)-based method that allows characterization of the structure and dynamics of biomolecular systems through providing quantitative experimental information on solvent accessibility of NMR-active nuclei. Addition of soluble paramagnetic probes to the solution of a biomolecule leads to paramagnetic relaxation enhancement in a concentration-dependent manner. Here we review recent progress in the sPRE-based characterization of structural and dynamic properties of biomolecules and their complexes, and aim to deliver a comprehensive illustration of a growing number of applications of the method to various biological systems. We discuss the physical principles of sPRE measurements and provide an overview of available co-solute paramagnetic probes. We then explore how sPRE, in combination with complementary biophysical techniques, can further advance biomolecular structure determination, identification of interaction surfaces within protein complexes, and probing of conformational changes and low-population transient states, as well as deliver insights into weak, nonspecific, and transient interactions between proteins and co-solutes. In addition, we present examples of how the incorporation of solvent paramagnetic probes can improve the sensitivity of NMR experiments and discuss the prospects of applying sPRE to NMR metabolomics, drug discovery, and the study of intrinsically disordered proteins.
Collapse
Affiliation(s)
- Aneta J Lenard
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Research Unit Integrative Structural Biology, Medical University of Graz, 8010 Graz, Austria.
| | - Frans A A Mulder
- Interdisciplinary Nanoscience Center and Department of Chemistry, University of Aarhus, DK-8000 Aarhus, Denmark; Institute of Biochemistry, Johannes Kepler Universität Linz, 4040 Linz, Austria.
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Ageing, Molecular Biology and Biochemistry, Research Unit Integrative Structural Biology, Medical University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria.
| |
Collapse
|
12
|
Pras A, Houben B, Aprile FA, Seinstra R, Gallardo R, Janssen L, Hogewerf W, Gallrein C, De Vleeschouwer M, Mata‐Cabana A, Koopman M, Stroo E, de Vries M, Louise Edwards S, Kirstein J, Vendruscolo M, Falsone SF, Rousseau F, Schymkowitz J, Nollen EAA. The cellular modifier MOAG-4/SERF drives amyloid formation through charge complementation. EMBO J 2021; 40:e107568. [PMID: 34617299 PMCID: PMC8561633 DOI: 10.15252/embj.2020107568] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 11/09/2022] Open
Abstract
While aggregation-prone proteins are known to accelerate aging and cause age-related diseases, the cellular mechanisms that drive their cytotoxicity remain unresolved. The orthologous proteins MOAG-4, SERF1A, and SERF2 have recently been identified as cellular modifiers of such proteotoxicity. Using a peptide array screening approach on human amyloidogenic proteins, we found that SERF2 interacted with protein segments enriched in negatively charged and hydrophobic, aromatic amino acids. The absence of such segments, or the neutralization of the positive charge in SERF2, prevented these interactions and abolished the amyloid-promoting activity of SERF2. In protein aggregation models in the nematode worm Caenorhabditis elegans, protein aggregation and toxicity were suppressed by mutating the endogenous locus of MOAG-4 to neutralize charge. Our data indicate that MOAG-4 and SERF2 drive protein aggregation and toxicity by interactions with negatively charged segments in aggregation-prone proteins. Such charge interactions might accelerate primary nucleation of amyloid by initiating structural changes and by decreasing colloidal stability. Our study points at charge interactions between cellular modifiers and amyloidogenic proteins as potential targets for interventions to reduce age-related protein toxicity.
Collapse
Affiliation(s)
- Anita Pras
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Bert Houben
- VIB‐KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- Switch LaboratoryDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Francesco A Aprile
- Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeCambridgeUK
- Present address:
Department of ChemistryMolecular Sciences Research HubImperial College LondonLondonUK
| | - Renée Seinstra
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Rodrigo Gallardo
- VIB‐KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- Switch LaboratoryDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
- Present address:
Astbury Centre for Structural Molecular BiologySchool of Molecular and Cellular BiologyUniversity of LeedsLeedsUK
| | - Leen Janssen
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Wytse Hogewerf
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Christian Gallrein
- Department of Molecular Physiology and Cell BiologyLeibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V. (FMP)BerlinGermany
| | - Matthias De Vleeschouwer
- VIB‐KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- Switch LaboratoryDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Alejandro Mata‐Cabana
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Mandy Koopman
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Esther Stroo
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Minke de Vries
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Samantha Louise Edwards
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Janine Kirstein
- Department of Molecular Physiology and Cell BiologyLeibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V. (FMP)BerlinGermany
- Faculty of Biology & ChemistryUniversity of BremenBremenGermany
| | - Michele Vendruscolo
- Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeCambridgeUK
| | | | - Frederic Rousseau
- VIB‐KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- Switch LaboratoryDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Joost Schymkowitz
- VIB‐KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- Switch LaboratoryDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Ellen A A Nollen
- European Research Institute for the Biology of AgeingUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| |
Collapse
|
13
|
Cleverley K, Lee WC, Mumford P, Collins T, Rickman M, Cunningham TJ, Cleak J, Mianne J, Szoke-Kovacs Z, Stewart M, Teboul L, Maduro C, Wells S, Wiseman FK, Fisher EMC. A novel knockout mouse for the small EDRK-rich factor 2 (Serf2) showing developmental and other deficits. Mamm Genome 2021; 32:94-103. [PMID: 33713180 PMCID: PMC8012326 DOI: 10.1007/s00335-021-09864-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/22/2021] [Indexed: 11/29/2022]
Abstract
The small EDRK-rich factor 2 (SERF2) is a highly conserved protein that modifies amyloid fibre assembly in vitro and promotes protein misfolding. However, the role of SERF2 in regulating age-related proteotoxicity remains largely unexplored due to a lack of in vivo models. Here, we report the generation of Serf2 knockout mice using an ES cell targeting approach, with Serf2 knockout alleles being bred onto different defined genetic backgrounds. We highlight phenotyping data from heterozygous Serf2+/− mice, including unexpected male-specific phenotypes in startle response and pre-pulse inhibition. We report embryonic lethality in Serf2−/− null animals when bred onto a C57BL/6 N background. However, homozygous null animals were viable on a mixed genetic background and, remarkably, developed without obvious abnormalities. The Serf2 knockout mice provide a powerful tool to further investigate the role of SERF2 protein in previously unexplored pathophysiological pathways in the context of a whole organism.
Collapse
Affiliation(s)
- Karen Cleverley
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK
| | - Weaverly Colleen Lee
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK
| | - Paige Mumford
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK.,The UK Dementia Research Institute, University College London, Queen Square, London, WC1N 3BG, UK
| | - Toby Collins
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK
| | - Matthew Rickman
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK
| | | | | | - Joffrey Mianne
- Mary Lyon Centre, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | | | | | | | - Cheryl Maduro
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK
| | - Sara Wells
- Mary Lyon Centre, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Frances K Wiseman
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK.,The UK Dementia Research Institute, University College London, Queen Square, London, WC1N 3BG, UK
| | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
14
|
Pras A, Nollen EAA. Regulation of Age-Related Protein Toxicity. Front Cell Dev Biol 2021; 9:637084. [PMID: 33748125 PMCID: PMC7973223 DOI: 10.3389/fcell.2021.637084] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/10/2021] [Indexed: 12/23/2022] Open
Abstract
Proteome damage plays a major role in aging and age-related neurodegenerative diseases. Under healthy conditions, molecular quality control mechanisms prevent toxic protein misfolding and aggregation. These mechanisms include molecular chaperones for protein folding, spatial compartmentalization for sequestration, and degradation pathways for the removal of harmful proteins. These mechanisms decline with age, resulting in the accumulation of aggregation-prone proteins that are harmful to cells. In the past decades, a variety of fast- and slow-aging model organisms have been used to investigate the biological mechanisms that accelerate or prevent such protein toxicity. In this review, we describe the most important mechanisms that are required for maintaining a healthy proteome. We describe how these mechanisms decline during aging and lead to toxic protein misassembly, aggregation, and amyloid formation. In addition, we discuss how optimized protein homeostasis mechanisms in long-living animals contribute to prolonging their lifespan. This knowledge might help us to develop interventions in the protein homeostasis network that delay aging and age-related pathologies.
Collapse
Affiliation(s)
| | - Ellen A. A. Nollen
- Laboratory of Molecular Neurobiology of Ageing, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Probing Surfaces in Dynamic Protein Interactions. J Mol Biol 2020; 432:2949-2972. [DOI: 10.1016/j.jmb.2020.02.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 01/09/2023]
|
16
|
Meyer NH, Dellago H, Tam-Amersdorfer C, Merle DA, Parlato R, Gesslbauer B, Almer J, Gschwandtner M, Leon A, Franzmann TM, Grillari J, Kungl AJ, Zangger K, Falsone SF. Structural Fuzziness of the RNA-Organizing Protein SERF Determines a Toxic Gain-of-interaction. J Mol Biol 2019; 432:930-951. [PMID: 31794729 DOI: 10.1016/j.jmb.2019.11.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022]
Abstract
The mechanisms by which protein complexes convert from functional to pathogenic are the subject of intensive research. Here, we report how functionally unfavorable protein interactions can be induced by structural fuzziness, i.e., by persisting conformational disorder in protein complexes. We show that extreme disorder in the bound state transforms the intrinsically disordered protein SERF1a from an RNA-organizing factor into a pathogenic enhancer of alpha-synuclein (aSyn) amyloid toxicity. We demonstrate that SERF1a promotes the incorporation of RNA into nucleoli and liquid-like artificial RNA-organelles by retaining an unusually high degree of conformational disorder in the RNA-bound state. However, this type of structural fuzziness also determines an undifferentiated interaction with aSyn. RNA and aSyn both bind to one identical, positively charged site of SERF1a by an analogous electrostatic binding mode, with similar binding affinities, and without any observable disorder-to-order transition. The absence of primary or secondary structure discriminants results in SERF1a being unable to select between nucleic acid and amyloidogenic protein, leading the pro-amyloid aSyn:SERF1a interaction to prevail in the cytosol under conditions of cellular stress. We suggest that fuzzy disorder in SERF1a complexes accounts for an adverse gain-of-interaction which favors toxic binding to aSyn at the expense of nontoxic RNA binding, thereby leading to a functionally distorted and pathogenic process. Thus, structural fuzziness constitutes a direct link between extreme conformational flexibility, amyloid aggregation, and the malfunctioning of RNA-associated cellular processes, three signatures of neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- N Helge Meyer
- Division of Experimental Allergology and Immunodermatology, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Hanna Dellago
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Wien, Austria
| | - Carmen Tam-Amersdorfer
- Institute of Pathophysiology and Immunology, Medical University of Graz, Heinrichstr. 31, 8010 Graz, Austria
| | - David A Merle
- Department of Ophthalmology, Medical University of Graz, Auenbruggerplatz 4, 8036 Graz, Austria
| | - Rosanna Parlato
- Institute of Applied Physiology, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany; Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Bernd Gesslbauer
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstr. 1, 8010 Graz, Austria
| | - Johannes Almer
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstr. 1, 8010 Graz, Austria
| | - Martha Gschwandtner
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstr. 1, 8010 Graz, Austria
| | - A Leon
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstr. 1, 8010 Graz, Austria
| | - Titus M Franzmann
- Biotechnology Center of the TU Dresden, Tatzberg 47/49, 01307 Dresden, Germany
| | - Johannes Grillari
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Wien, Austria; Ludwig Boltzmann Institute of Experimental and Clinical Traumatology, Donaueschingenstr. 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Andreas J Kungl
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstr. 1, 8010 Graz, Austria
| | - Klaus Zangger
- Institute of Chemistry, University of Graz, Heinrichstr. 28, 8010 Graz, Austria
| | - S Fabio Falsone
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstr. 1, 8010 Graz, Austria; Steiermärkische Krankenanstaltengesellschaft m.b.H. (KAGes), Stiftingtalstraße 4-6, 8010, Graz, Austria.
| |
Collapse
|
17
|
SERF engages in a fuzzy complex that accelerates primary nucleation of amyloid proteins. Proc Natl Acad Sci U S A 2019; 116:23040-23049. [PMID: 31659041 DOI: 10.1073/pnas.1913316116] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The assembly of small disordered proteins into highly ordered amyloid fibrils in Alzheimer's and Parkinson's patients is closely associated with dementia and neurodegeneration. Understanding the process of amyloid formation is thus crucial in the development of effective treatments for these devastating neurodegenerative diseases. Recently, a tiny, highly conserved and disordered protein called SERF was discovered to modify amyloid formation in Caenorhabditis elegans and humans. Here, we use kinetics measurements and native ion mobility-mass spectrometry to show that SERF mainly affects the rate of primary nucleation in amyloid formation for the disease-related proteins Aβ40 and α-synuclein. SERF's high degree of plasticity enables it to bind various conformations of monomeric Aβ40 and α-synuclein to form structurally diverse, fuzzy complexes. This structural diversity persists into early stages of amyloid formation. Our results suggest that amyloid nucleation is considerably more complex than age-related conversion of Aβ40 and α-synuclein into single amyloid-prone conformations.
Collapse
|
18
|
Wong ETC, Gsponer J. Predicting Protein-Protein Interfaces that Bind Intrinsically Disordered Protein Regions. J Mol Biol 2019; 431:3157-3178. [PMID: 31207240 DOI: 10.1016/j.jmb.2019.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Abstract
A long-standing goal in biology is the complete annotation of function and structure on all protein-protein interactions, a large fraction of which is mediated by intrinsically disordered protein regions (IDRs). However, knowledge derived from experimental structures of such protein complexes is disproportionately small due, in part, to challenges in studying interactions of IDRs. Here, we introduce IDRBind, a computational method that by combining gradient boosted trees and conditional random field models predicts binding sites of IDRs with performance approaching state-of-the-art globular interface predictions, making it suitable for proteome-wide applications. Although designed and trained with a focus on molecular recognition features, which are long interaction-mediating-elements in IDRs, IDRBind also predicts the binding sites of short peptides more accurately than existing specialized predictors. Consistent with IDRBind's specificity, a comparison of protein interface categories uncovered uniform trends in multiple physicochemical properties, positioning molecular recognition feature interfaces between peptide and globular interfaces.
Collapse
Affiliation(s)
- Eric T C Wong
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Jörg Gsponer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|