1
|
Lambert-Smith IA, Shephard VK, McAlary L, Yerbury JJ, Saunders DN. High-content analysis of proteostasis capacity in cellular models of amyotrophic lateral sclerosis (ALS). Sci Rep 2024; 14:13844. [PMID: 38879591 PMCID: PMC11180180 DOI: 10.1038/s41598-024-64366-0] [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: 11/22/2021] [Accepted: 06/07/2024] [Indexed: 06/19/2024] Open
Abstract
Disrupted proteome homeostasis (proteostasis) in amyotrophic lateral sclerosis (ALS) has been a major focus of research in the past two decades. However, the proteostasis processes that become disturbed in ALS are not fully understood. Obtaining more detailed knowledge of proteostasis disruption in association with different ALS-causing mutations will improve our understanding of ALS pathophysiology and may identify novel therapeutic targets and strategies for ALS patients. Here we describe the development and use of a novel high-content analysis (HCA) assay to investigate proteostasis disturbances caused by the expression of several ALS-causing gene variants. This assay involves the use of conformationally-destabilised mutants of firefly luciferase (Fluc) to examine protein folding/re-folding capacity in NSC-34 cells expressing ALS-associated mutations in the genes encoding superoxide dismutase-1 (SOD1A4V) and cyclin F (CCNFS621G). We demonstrate that these Fluc isoforms can be used in high-throughput format to report on reductions in the activity of the chaperone network that result from the expression of SOD1A4V, providing multiplexed information at single-cell resolution. In addition to SOD1A4V and CCNFS621G, NSC-34 models of ALS-associated TDP-43, FUS, UBQLN2, OPTN, VCP and VAPB mutants were generated that could be screened using this assay in future work. For ALS-associated mutant proteins that do cause reductions in protein quality control capacity, such as SOD1A4V, this assay has potential to be applied in drug screening studies to identify candidate compounds that can ameliorate this deficiency.
Collapse
Affiliation(s)
- Isabella A Lambert-Smith
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia.
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia.
| | - Victoria K Shephard
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Luke McAlary
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Darren N Saunders
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
2
|
Abidi SMS, Shukla AK, Randhawa S, Bathla M, Acharya A. Diosgenin loaded cellulose nanoonion impedes different stages of protein aggregation induced cell death via alleviating mitochondrial dysfunction and upregulation of autophagy. Int J Biol Macromol 2024; 266:131108. [PMID: 38531523 DOI: 10.1016/j.ijbiomac.2024.131108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 03/28/2024]
Abstract
Protein aggregation is a multifaceted phenomenon prevalent in the progression of neurodegenerative diseases, yielding aggregates of diverse sizes. Recently, increased attention has been directed towards early protein aggregates due to their pronounced toxicity, largely stemming from inflammation mediated by reactive oxygen species (ROS). This study advocates for a therapeutic approach focusing on inflammation control rather than mere ROS inhibition in the context of neurodegenerative disorders. Here, we introduced Camellia sinensis cellulose nanoonion (CS-CNO) as an innovative, biocompatible nanocarrier for encapsulating the phytosteroid diosgenin (DGN@CS-CNO). The resulting nano-assembly, manifesting as spherical entities with dimensions averaging ~180-220 nm, exhibits a remarkable capacity for the gradual and sustained release of approximately 39-44 % of DGN over a 60-hour time frame. DGN@CS-CNO displays a striking ability to inhibit or disassemble various phases of hen egg white lysozyme (HEWL) protein aggregates, including the early (HEWLEA) and late (HEWLLA) stages. In vitro experiments employing HEK293 cells underscore the potential of DGN@CS-CNO in mitigating cell death provoked by protein aggregation. This effect is achieved by ameliorating ROS-mediated inflammation and countering mitochondrial dysfunction, as evidenced by alterations in TNFα, TLR4, and MT-CO1 protein expression. Western blot analyses reveal that the gradual and sustained release of DGN from DGN@CS-CNO induces autophagy, a pivotal process in dismantling intracellular amyloid deposits. In summary, this study not only illuminates a path forward but also presents a compelling case for the utilization of phytosteroid as a formidable strategy against neuroinflammation incited by protein aggregation.
Collapse
Affiliation(s)
- Syed M S Abidi
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ashish K Shukla
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shiwani Randhawa
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manik Bathla
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
3
|
Mansuri S, Jain A, Singh R, Rawat S, Mondal D, Raychaudhuri S. Widespread nuclear lamina injuries defeat proteostatic purposes of α-synuclein amyloid inclusions. J Cell Sci 2024; 137:jcs261935. [PMID: 38477372 DOI: 10.1242/jcs.261935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Biogenesis of inclusion bodies (IBs) facilitates protein quality control (PQC). Canonical aggresomes execute degradation of misfolded proteins while non-degradable amyloids sequester into insoluble protein deposits. Lewy bodies (LBs) are filamentous amyloid inclusions of α-synuclein, but PQC benefits and drawbacks associated with LB-like IBs remain underexplored. Here, we report that crosstalk between filamentous LB-like IBs and aggresome-like IBs of α-synuclein (Syn-aggresomes) buffer the load, aggregation state, and turnover of the amyloidogenic protein in mouse primary neurons and HEK293T cells. Filamentous LB-like IBs possess unorthodox PQC capacities of self-quarantining α-synuclein amyloids and being degradable upon receding fresh amyloidogenesis. Syn-aggresomes equilibrate biogenesis of filamentous LB-like IBs by facilitating spontaneous degradation of α-synuclein and conditional turnover of disintegrated α-synuclein amyloids. Thus, both types of IB primarily contribute to PQC. Incidentally, the overgrown perinuclear LB-like IBs become degenerative once these are misidentified by BICD2, a cargo-adapter for the cytosolic motor-protein dynein. Microscopy indicates that microtubules surrounding the perinuclear filamentous inclusions are also distorted, misbalancing the cytoskeleton-nucleoskeleton tension leading to widespread lamina injuries. Together, nucleocytoplasmic mixing, DNA damage, and deregulated transcription of stress chaperones defeat the proteostatic purposes of the filamentous amyloids of α-synuclein.
Collapse
Affiliation(s)
- Shemin Mansuri
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
| | - Aanchal Jain
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Richa Singh
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
| | - Shivali Rawat
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
| | - Debodyuti Mondal
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
| | - Swasti Raychaudhuri
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
4
|
Iyer AK, Schoch KM, Verbeck A, Galasso G, Chen H, Smith S, Oldenborg A, Miller TM, Karch CM, Bonni A. Targeted ASO-mediated Atp1a2 knockdown in astrocytes reduces SOD1 aggregation and accelerates disease onset in mutant SOD1 mice. PLoS One 2023; 18:e0294731. [PMID: 38015828 PMCID: PMC10683999 DOI: 10.1371/journal.pone.0294731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023] Open
Abstract
Astrocyte-specific ion pump α2-Na+/K+-ATPase plays a critical role in the pathogenesis of amyotrophic lateral sclerosis (ALS). Here, we test the effect of Atp1a2 mRNA-specific antisense oligonucleotides (ASOs) to induce α2-Na+/K+-ATPase knockdown in the widely used ALS animal model, SOD1*G93A mice. Two ASOs led to efficient Atp1a2 knockdown and significantly reduced SOD1 aggregation in vivo. Although Atp1a2 ASO-treated mice displayed no off-target or systemic toxicity, the ASO-treated mice exhibited an accelerated disease onset and shorter lifespan than control mice. Transcriptomics studies reveal downregulation of genes involved in oxidative response, metabolic pathways, trans-synaptic signaling, and upregulation of genes involved in glutamate receptor signaling and complement activation, suggesting a potential role for these molecular pathways in de-coupling SOD1 aggregation from survival in Atp1a2 ASO-treated mice. Together, these results reveal a role for α2-Na+/K+-ATPase in SOD1 aggregation and highlight the critical effect of temporal modulation of genetically validated therapeutic targets in neurodegenerative diseases.
Collapse
Affiliation(s)
- Abhirami K. Iyer
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kathleen M. Schoch
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Anthony Verbeck
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Grant Galasso
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Hao Chen
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sarah Smith
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Anna Oldenborg
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Timothy M. Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Azad Bonni
- Neuroscience and Rare Diseases, Roche Pharma Research and Early Development (pRED), Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
5
|
Nowakowska AW, Wojciechowski JW, Szulc N, Kotulska M. The role of tandem repeats in bacterial functional amyloids. J Struct Biol 2023; 215:108002. [PMID: 37482232 DOI: 10.1016/j.jsb.2023.108002] [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: 03/24/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
Repetitivity and modularity of proteins are two related notions incorporated into multiple evolutionary concepts. We discuss whether they may also be essential for functional amyloids. Amyloids are proteins that create very regular and usually highly insoluble fibrils, which are often associated with neurodegeneration. However, recent discoveries showed that amyloid structure of a protein could also be beneficial and desired, e.g., to promote cell adhesion. Functional amyloids are proteins which differ in their characteristics from pathological amyloids, so that the fibril formation could be more under control of an organism. We propose that repeats in the sequence could regulate the aggregation propensity of these proteins. The inclusion of multiple symmetric interactions, due to the presence of the repeats, could be supporting and strengthening the desirable structural properties of functional amyloids. Our results show that tandem repeats in bacterial functional amyloids have a distinct characteristic. The pattern of repeats supports the appropriate level of fibril formation and better controllability of fibril stability. The repeats tend to be more imperfect, which attenuates excessive aggregation propensity. Their desired structure and function are also reinforced by their amino acid profile. Although in the study we focused on bacterial functional amyloids, due to their importance in biofilm formation, we propose that similar mechanisms could be employed in other functional amyloids which are designed by evolution to aggregate in a desirable manner, but not necessarily in pathological amyloids.
Collapse
Affiliation(s)
- Alicja W Nowakowska
- Wrocław University of Science and Technology, Department of Biomedical Engineering, Poland.
| | - Jakub W Wojciechowski
- Wrocław University of Science and Technology, Department of Biomedical Engineering, Poland
| | - Natalia Szulc
- Wrocław University of Science and Technology, Department of Biomedical Engineering, Poland; Wrocław University of Environmental and Life Sciences, Department of Physics and Biophysics, Poland; LPCT, CNRS, Universite de Lorraine, F-54000 Nancy, France
| | - Malgorzata Kotulska
- Wrocław University of Science and Technology, Department of Biomedical Engineering, Poland.
| |
Collapse
|
6
|
Maity A, Mondal A, Kundu S, Shome G, Misra R, Singh A, Pal U, Mandal AK, Bera K, Maiti NC. Naringenin-Functionalized Gold Nanoparticles and Their Role in α-Synuclein Stabilization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:7231-7248. [PMID: 37094111 DOI: 10.1021/acs.langmuir.2c03259] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Misfolding and self-assembly of several intrinsically disordered proteins into ordered β-sheet-rich amyloid aggregates emerged as hallmarks of several neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Here we show how the naringenin-embedded nanostructure effectively retards aggregation and fibril formation of α-synuclein, which is strongly associated with the pathology of Parkinson's-like diseases. Naringenin is a polyphenolic compound from a plant source, and in our current investigation, we reported the one-pot synthesis of naringenin-coated spherical and monophasic gold nanoparticles (NAR-AuNPs) under optimized conditions. The average hydrodynamic diameter of the produced nanoparticle was ∼24 nm and showed a distinct absorption band at 533 nm. The zeta potential of the nanocomposite was ∼-22 mV and indicated the presence of naringenin on the surface of nanoparticles. Core-level XPS spectrum analysis showed prominent peaks at 84.02 and 87.68 eV, suggesting the zero oxidation state of metal in the nanostructure. Additionally, the peaks at 86.14 and 89.76 eV were due to the Au-O bond, induced by the hydroxyl groups of the naringenin molecule. The FT-IR analysis further confirmed strong interactions of the molecule with the gold nanosurface via the phenolic oxygen group. The composite surface was found to interact with monomeric α-synuclein and caused a red shift in the nanoparticle absorption band by ∼5 nm. The binding affinity of the composite nanostructure toward α-synuclein was in the micromolar range (Ka∼ 5.02 × 106 M-1) and may produce a protein corona over the gold nanosurface. A circular dichroism study showed that the nanocomposite can arrest the conformational fluctuation of the protein and hindered its transformation into a compact cross-β-sheet conformation, a prerequisite for amyloid fibril formation. Furthermore, it was found that naringenin and its nanocomplex did not perturb the viability of neuronal cells. It thus appeared that engineering of the nanosurface with naringenin could be an alternative strategy in developing treatment approaches for Parkinson's and other diseases linked to protein conformation.
Collapse
Affiliation(s)
- Anupam Maity
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Animesh Mondal
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Shubham Kundu
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Gourav Shome
- Division of Molecular Medicine, Bose Institute, Kolkata 700091, India
| | - Rajdip Misra
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Aakriti Singh
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Uttam Pal
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Atin Kumar Mandal
- Division of Molecular Medicine, Bose Institute, Kolkata 700091, India
| | - Kaushik Bera
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
- Department of Chemistry, The Heritage School, 994 Chowbaga Road, Anandapur, East Kolkata Twp, Kolkata 700107, India
| | - Nakul C Maiti
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| |
Collapse
|
7
|
Aggregation of Irisin and its Prevention by Trehalose: A Biophysical Approach. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
8
|
Henderson RD, Kepp KP, Eisen A. ALS/FTD: Evolution, Aging, and Cellular Metabolic Exhaustion. Front Neurol 2022; 13:890203. [PMID: 35711269 PMCID: PMC9196861 DOI: 10.3389/fneur.2022.890203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD) are neurodegenerations with evolutionary underpinnings, expansive clinical presentations, and multiple genetic risk factors involving a complex network of pathways. This perspective considers the complex cellular pathology of aging motoneuronal and frontal/prefrontal cortical networks in the context of evolutionary, clinical, and biochemical features of the disease. We emphasize the importance of evolution in the development of the higher cortical function, within the influence of increasing lifespan. Particularly, the role of aging on the metabolic competence of delicately optimized neurons, age-related increased proteostatic costs, and specific genetic risk factors that gradually reduce the energy available for neuronal function leading to neuronal failure and disease.
Collapse
Affiliation(s)
| | - Kasper Planeta Kepp
- Department of Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Andrew Eisen
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
9
|
Lambert-Smith IA, Saunders DN, Yerbury JJ. Progress in biophysics and molecular biology proteostasis impairment and ALS. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 174:3-27. [PMID: 35716729 DOI: 10.1016/j.pbiomolbio.2022.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 05/19/2022] [Accepted: 06/09/2022] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disease that results from the loss of both upper and lower motor neurons. It is the most common motor neuron disease and currently has no effective treatment. There is mounting evidence to suggest that disturbances in proteostasis play a significant role in ALS pathogenesis. Proteostasis is the maintenance of the proteome at the right level, conformation and location to allow a cell to perform its intended function. In this review, we present a thorough synthesis of the literature that provides evidence that genetic mutations associated with ALS cause imbalance to a proteome that is vulnerable to such pressure due to its metastable nature. We propose that the mechanism underlying motor neuron death caused by defects in mRNA metabolism and protein degradation pathways converges on proteostasis dysfunction. We propose that the proteostasis network may provide an effective target for therapeutic development in ALS.
Collapse
Affiliation(s)
- Isabella A Lambert-Smith
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Darren N Saunders
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
10
|
Wang Y, Wu C. The effect of mechanical shocks on the initial aggregation behavior of yeast prion protein Sup35NM. Int J Biol Macromol 2022; 212:465-473. [PMID: 35618091 DOI: 10.1016/j.ijbiomac.2022.05.127] [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: 02/24/2022] [Revised: 05/05/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022]
Abstract
To study the effect of mechanical shocks on the neurodegenerative-related fibril-formation protein, the aggregation process, especially the initial oligomerization of a model yeast prion protein Sup35NM, was followed and analyzed by using a combination of laser light scattering, the Smoluchowski coagulation analysis, Thioflavin T fluorescence assay, and transmission electron microscopy. We find that an initial short-time mechanical shock (ultrasonication or circular shaking) affects the in vitro association kinetics of neurodegenerative-related Sup35NM proteins in dilute PBS solutions by generating a relatively larger number of smaller non-structured oligomers that further serve as tiny "crystallization" seeds in promoting the formation of longer fibrils. Our study provides an effective and quantitative method to investigate the initial oligomerization kinetics of amyloid fibrils formation. Furthermore, the current results may shed light on the molecular understanding on how environmental factors increase the risk of neurodegenerative diseases such as dementia.
Collapse
Affiliation(s)
- Yanjing Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Chi Wu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China; Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China; Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemical Physics, The University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
11
|
Constructing conformational library for amyloid-β42 dimers as the smallest toxic oligomers using two CHARMM force fields. J Mol Graph Model 2022; 115:108207. [DOI: 10.1016/j.jmgm.2022.108207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/19/2022]
|
12
|
Zielinski M, Röder C, Schröder GF. Challenges in sample preparation and structure determination of amyloids by cryo-EM. J Biol Chem 2021; 297:100938. [PMID: 34224730 PMCID: PMC8335658 DOI: 10.1016/j.jbc.2021.100938] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 01/12/2023] Open
Abstract
Amyloids share a common architecture but play disparate biological roles in processes ranging from bacterial defense mechanisms to protein misfolding diseases. Their structures are highly polymorphic, which makes them difficult to study by X-ray diffraction or NMR spectroscopy. Our understanding of amyloid structures is due in large part to recent advances in the field of cryo-EM, which allows for determining the polymorphs separately. In this review, we highlight the main stepping stones leading to the substantial number of high-resolution amyloid fibril structures known today as well as recent developments regarding automation and software in cryo-EM. We discuss that sample preparation should move closer to physiological conditions to understand how amyloid aggregation and disease are linked. We further highlight new approaches to address heterogeneity and polymorphism of amyloid fibrils in EM image processing and give an outlook to the upcoming challenges in researching the structural biology of amyloids.
Collapse
Affiliation(s)
- Mara Zielinski
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany
| | - Christine Röder
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany; Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Gunnar F Schröder
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany; Physics Department, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
13
|
Ahmadi H, Shogen K, Fujita K, Honjo T, Kakimi K, Futami J. Unusual aggregation property of recombinantly expressed cancer-testis antigens in mammalian cells. J Biochem 2021; 170:435-443. [PMID: 34247245 DOI: 10.1093/jb/mvab081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/02/2021] [Indexed: 11/13/2022] Open
Abstract
Transient expression of human intracellular proteins in human embryonic kidney (HEK) 293 cells is a reliable system for obtaining soluble proteins with biologically active conformations. Contrary to conventional concepts, we found that recombinantly expressed intracellular cancer-testis antigens (CTAs) showed frequent aggregation in HEK293 cells. Although experimental subcellular localization of recombinant CTAs displayed proper cytosolic or nuclear localization, some proteins showed aggregated particles in the cell. This aggregative property was not observed in recombinant housekeeping proteins. No significant correlation was found between the aggregative and biophysical properties, such as hydrophobicity, contents of intrinsically disordered regions, and expression levels, of CTAs. These results can be explained in terms of structural instability of CTAs, which are specifically expressed in the testis and aberrantly expressed in cancer cells and function as a hub in the protein-protein network using intrinsically disordered regions. Hence, we speculate that recombinantly expressed CTAs failed to form this protein complex. Thus, unfolded CTAs formed aggregated particles in the cell.
Collapse
Affiliation(s)
- Hannaneh Ahmadi
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Kohei Shogen
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| | - Kana Fujita
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| | - Tomoko Honjo
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Junichiro Futami
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.,Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| |
Collapse
|
14
|
Wells C, Brennan S, Keon M, Ooi L. The role of amyloid oligomers in neurodegenerative pathologies. Int J Biol Macromol 2021; 181:582-604. [PMID: 33766600 DOI: 10.1016/j.ijbiomac.2021.03.113] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/18/2021] [Accepted: 03/19/2021] [Indexed: 11/25/2022]
Abstract
Many neurodegenerative diseases are rooted in the activities of amyloid-like proteins which possess conformations that spread to healthy proteins. These include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS). While their clinical manifestations vary, their protein-level mechanisms are remarkably similar. Aberrant monomeric proteins undergo conformational shifts, facilitating aggregation and formation of solid fibrils. However, there is growing evidence that intermediate oligomeric stages are key drivers of neuronal toxicity. Analysis of protein dynamics is complicated by the fact that nucleation and growth of amyloid-like proteins is not a linear pathway. Feedback within this pathway results in exponential acceleration of aggregation, but activities exerted by oligomers and fibrils can alter cellular interactions and the cellular environment as a whole. The resulting cascade of effects likely contributes to the late onset and accelerating progression of amyloid-like protein disorders and the widespread effects they have on the body. In this review we explore the amyloid-like proteins associated with AD, PD, HD and ALS, as well as the common mechanisms of amyloid-like protein nucleation and aggregation. From this, we identify core elements of pathological progression which have been targeted for therapies, and which may become future therapeutic targets.
Collapse
Affiliation(s)
- Cameron Wells
- GenieUs Genomics, Sydney, NSW 2010, Australia; University of New South Wales, Sydney, NSW 2052, Australia
| | | | - Matt Keon
- GenieUs Genomics, Sydney, NSW 2010, Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Chemistry and Molecular Bioscience, and Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia; GenieUs Genomics, Sydney, NSW 2010, Australia
| |
Collapse
|
15
|
Trehalose Restrains the Fibril Load towards α-Lactalbumin Aggregation and Halts Fibrillation in a Concentration-Dependent Manner. Biomolecules 2021; 11:biom11030414. [PMID: 33799517 PMCID: PMC8001226 DOI: 10.3390/biom11030414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 01/12/2023] Open
Abstract
Protein aggregation and misfolding are some of the most challenging obstacles, customarily studied for their association with amyloid pathologies. The mechanism of amyloid fibrillation development is a dynamic phenomenon involving various factors such as the intrinsic properties of protein and the physical and chemical environmental conditions. The purpose of this study was to see the thermal aggregation profile of alpha-lactalbumin (α-LA) and to delineate the effect of trehalose on its aggregation profile. α-LA was subjected to thermal aggregation at high concentrations. UV-Vis spectroscopy, a turbidity assay, intrinsic fluorescence, Rayleigh scattering and a thioflavin T (ThT) assay explained the steady outcomes that 1 M trehalose repressed α-LA aggregation in the most effective way followed by 0.75 M and 0.5 M and to a significantly lesser degree by 0.25 M. Multi spectroscopic obser Sania Bashir ations were further entrenched by microscopy. Transmission electron microscopy confirmed that in the presence of its higher concentration, trehalose hinders fibril development in α-LA. In vitro studies were further validated by in silico studies. Molecular docking analysis indicated that trehalose occupied the binding pocket cavity of α-LA and offered several significant interactions, including H-bonds with important residues. This study provides a platform for trehalose in the therapeutic management of protein aggregation-related diseases.
Collapse
|
16
|
Kim S, Kwon M, Hwang Y, Yoon J, Park S, Kang HC. Stress-induced NEDDylation promotes cytosolic protein aggregation through HDAC6 in a p62-dependent manner. iScience 2021; 24:102146. [PMID: 33665565 PMCID: PMC7903351 DOI: 10.1016/j.isci.2021.102146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/07/2021] [Accepted: 02/01/2021] [Indexed: 02/08/2023] Open
Abstract
Stress-coupled NEDDylation potentially regulates the aggregation of nuclear proteins, which could protect the nuclear ubiquitin-proteasome system from proteotoxic stress. However, it remains unclear how NEDDylation controls protein-aggregation responses to diverse stress conditions. Here, we identified HDAC6 as a direct NEDD8-binding partner that regulates the formation of aggresome-like bodies (ALBs) containing NEDDylated cytosolic protein aggregates during ubiquitin stress. HDAC6 colocalizes with stress-induced ALBs, and HDAC6 inhibition suppresses ALBs formation, but not stress-induced NEDDylation, suggesting that HDAC6 carries NEDDylated-proteins to generate ALBs. Then, we monitored the ALBs-associated proteostasis network and found that p62 directly controls ALBs formation as an acceptor of NEDDylated cytosolic aggregates. Interestingly, we also observed that ALBs are highly condensed in chloroquine-treated cells with impaired autophagic flux, indicating that ALBs rely on autophagy. Collectively, our data suggest that NEDD8, HDAC6, and p62 are involved in the management of proteotoxic stress by forming cytosolic ALBs coupled to the aggresome-autophagy flux. NEDD8 directly binds to HDAC6 and regulates the formation of aggresome-like body (ALB) HDAC6 carries NEDDylated cytosolic protein aggregates into ALBs under ubiquitin stress p62 directly controls ALBs formation as an acceptor of NEDDylated cytosolic aggregates The NEDD8-HDAC6-p62 axis controls proteostasis by forming ALB-coupled autophagy
Collapse
Affiliation(s)
- Soyeon Kim
- Department of Physiology, Ajou University School of Medicine, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea
| | - Mira Kwon
- Department of Physiology, Ajou University School of Medicine, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea
| | - Yiseul Hwang
- Department of Physiology, Ajou University School of Medicine, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea
| | - Junghyun Yoon
- Department of Physiology, Ajou University School of Medicine, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea
| | - Sangwook Park
- Department of Physiology, Ajou University School of Medicine, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea
| | - Ho Chul Kang
- Department of Physiology, Ajou University School of Medicine, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499, Republic of Korea
| |
Collapse
|
17
|
Kabir MT, Uddin MS, Mathew B, Das PK, Perveen A, Ashraf GM. Emerging Promise of Immunotherapy for Alzheimer's Disease: A New Hope for the Development of Alzheimer's Vaccine. Curr Top Med Chem 2021; 20:1214-1234. [PMID: 32321405 DOI: 10.2174/1568026620666200422105156] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the characteristics of this devastating disorder include the progressive and disabling deficits in the cognitive functions including reasoning, attention, judgment, comprehension, memory, and language. OBJECTIVE In this article, we have focused on the recent progress that has been achieved in the development of an effective AD vaccine. SUMMARY Currently, available treatment options of AD are limited to deliver short-term symptomatic relief only. A number of strategies targeting amyloid-beta (Aβ) have been developed in order to treat or prevent AD. In order to exert an effective immune response, an AD vaccine should contain adjuvants that can induce an effective anti-inflammatory T helper 2 (Th2) immune response. AD vaccines should also possess the immunogens which have the capacity to stimulate a protective immune response against various cytotoxic Aβ conformers. The induction of an effective vaccine's immune response would necessitate the parallel delivery of immunogen to dendritic cells (DCs) and their priming to stimulate a Th2-polarized response. The aforesaid immune response is likely to mediate the generation of neutralizing antibodies against the neurotoxic Aβ oligomers (AβOs) and also anti-inflammatory cytokines, thus preventing the AD-related inflammation. CONCLUSION Since there is an age-related decline in the immune functions, therefore vaccines are more likely to prevent AD instead of providing treatment. AD vaccines might be an effective and convenient approach to avoid the treatment-related huge expense.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
18
|
Plotkin SS, Cashman NR. Passive immunotherapies targeting Aβ and tau in Alzheimer's disease. Neurobiol Dis 2020; 144:105010. [PMID: 32682954 PMCID: PMC7365083 DOI: 10.1016/j.nbd.2020.105010] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Amyloid-β (Aβ) and tau proteins currently represent the two most promising targets to treat Alzheimer's disease. The most extensively developed method to treat the pathologic forms of these proteins is through the administration of exogenous antibodies, or passive immunotherapy. In this review, we discuss the molecular-level strategies that researchers are using to design an effective therapeutic antibody, given the challenges in treating this disease. These challenges include selectively targeting a protein that has misfolded or is pathological rather than the more abundant, healthy protein, designing strategic constructs for immunizing an animal to raise an antibody that has the appropriate conformational selectivity to achieve this end, and clearing the pathological protein species before prion-like cell-to-cell spread of misfolded protein has irreparably damaged neurons, without invoking damaging inflammatory responses in the brain that naturally arise when the innate immune system is clearing foreign agents. The various solutions to these problems in current clinical trials will be discussed.
Collapse
Affiliation(s)
- Steven S Plotkin
- University of British Columbia, Department of Physics and Astronomy and Genome Sciences and Technology Program, Vancouver, BC V6T 1Z1, Canada.
| | - Neil R Cashman
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
19
|
Braun DJ, Dimayuga E, Morganti JM, Van Eldik LJ. Microglial-associated responses to comorbid amyloid pathology and hyperhomocysteinemia in an aged knock-in mouse model of Alzheimer's disease. J Neuroinflammation 2020; 17:274. [PMID: 32943069 PMCID: PMC7499995 DOI: 10.1186/s12974-020-01938-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Elevated blood homocysteine levels, termed hyperhomocysteinemia (HHcy), is a prevalent risk factor for Alzheimer's disease (AD) in elderly populations. While dietary supplementation of B-vitamins is a generally effective method to lower homocysteine levels, there is little if any benefit to cognition. In the context of amyloid pathology, dietary-induced HHcy is known to enhance amyloid deposition and certain inflammatory responses. Little is known, however, about whether there is a more specific effect on microglia resulting from combined amyloid and HHcy pathologies. METHODS The present study used a knock-in mouse model of amyloidosis, aged to 12 months, given 8 weeks of B-vitamin deficiency-induced HHcy to better understand how microglia are affected in this comorbidity context. RESULTS We found that HHcy-inducing diet increased amyloid plaque burden, altered the neuroinflammatory milieu, and upregulated the expression of multiple damage-associated and "homeostatic" microglial genes. CONCLUSIONS Taken together, these data indicate complex effects of comorbid pathologies on microglial function that are not driven solely by increased amyloid burden. Given the highly dynamic nature of microglia, their central role in AD pathology, and the frequent occurrence of various comorbidities in AD patients, it is increasingly important to understand how microglia respond to mixed pathological processes.
Collapse
Affiliation(s)
- David J Braun
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA.
| | - Edgardo Dimayuga
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
| | - Josh M Morganti
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA. .,Department of Neuroscience, University of Kentucky, Lexington, KY, USA. .,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
20
|
Foley AR, Raskatov JA. Assessing Reproducibility in Amyloid β Research: Impact of Aβ Sources on Experimental Outcomes. Chembiochem 2020; 21:2425-2430. [PMID: 32249510 PMCID: PMC7647053 DOI: 10.1002/cbic.202000125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/04/2020] [Indexed: 12/16/2022]
Abstract
The difficulty of synthesizing and purifying the amyloid β (Aβ) peptide, combined with its high aggregation propensity and low solubility under physiological conditions, leads to a wide variety of experimental results from kinetic assays to biological activity. Thus, it becomes challenging to reproduce outcomes, and this limits our ability to rely on reported results as the foundation for new research. This article examines variability of the Aβ peptide from different sources, comparing purity, and oligomer and fibril formation propensity side by side. The results highlight the importance of performing rigorous controls so that meaningful biophysical, biochemical, and neurobiological results can be obtained to improve our understanding on Aβ.
Collapse
Affiliation(s)
- Alejandro R Foley
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Jevgenij A Raskatov
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA
| |
Collapse
|
21
|
Babazadeh R, Ahmadpour D, Jia S, Hao X, Widlund P, Schneider K, Eisele F, Edo LD, Smits GJ, Liu B, Nystrom T. Syntaxin 5 Is Required for the Formation and Clearance of Protein Inclusions during Proteostatic Stress. Cell Rep 2020; 28:2096-2110.e8. [PMID: 31433985 DOI: 10.1016/j.celrep.2019.07.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 06/14/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
Spatial sorting to discrete quality control sites in the cell is a process harnessing the toxicity of aberrant proteins. We show that the yeast t-snare phosphoprotein syntaxin5 (Sed5) acts as a key factor in mitigating proteotoxicity and the spatial deposition and clearance of IPOD (insoluble protein deposit) inclusions associates with the disaggregase Hsp104. Sed5 phosphorylation promotes dynamic movement of COPII-associated Hsp104 and boosts disaggregation by favoring anterograde ER-to-Golgi trafficking. Hsp104-associated aggregates co-localize with Sed5 as well as components of the ER, trans Golgi network, and endocytic vesicles, transiently during proteostatic stress, explaining mechanistically how misfolded and aggregated proteins formed at the vicinity of the ER can hitchhike toward vacuolar IPOD sites. Many inclusions become associated with mitochondria in a HOPS/vCLAMP-dependent manner and co-localize with Vps39 (HOPS/vCLAMP) and Vps13, which are proteins providing contacts between vacuole and mitochondria. Both Vps39 and Vps13 are required also for efficient Sed5-dependent clearance of aggregates.
Collapse
Affiliation(s)
- Roja Babazadeh
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Doryaneh Ahmadpour
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Song Jia
- School of Life Science, Northeast Agricultural University, No. 600 Changjiang Street, Xiangfang District, Harbin 150030, China
| | - Xinxin Hao
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Per Widlund
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Kara Schneider
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Frederik Eisele
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Laura Dolz Edo
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1090, the Netherlands
| | - Gertien J Smits
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1090, the Netherlands
| | - Beidong Liu
- Department of Chemistry & Molecular Biology, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Thomas Nystrom
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg 405 30, Sweden.
| |
Collapse
|
22
|
Lackie RE, Marques-Lopes J, Ostapchenko VG, Good S, Choy WY, van Oosten-Hawle P, Pasternak SH, Prado VF, Prado MAM. Increased levels of Stress-inducible phosphoprotein-1 accelerates amyloid-β deposition in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2020; 8:143. [PMID: 32825842 PMCID: PMC7441634 DOI: 10.1186/s40478-020-01013-5] [Citation(s) in RCA: 8] [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: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 02/08/2023] Open
Abstract
Molecular chaperones and co-chaperones, which are part of the protein quality control machinery, have been shown to regulate distinct aspects of Alzheimer's Disease (AD) pathology in multiple ways. Notably, the co-chaperone STI1, which presents increased levels in AD, can protect mammalian neurons from amyloid-β toxicity in vitro and reduced STI1 levels worsen Aβ toxicity in C. elegans. However, whether increased STI1 levels can protect neurons in vivo remains unknown. We determined that overexpression of STI1 and/or Hsp90 protected C. elegans expressing Aβ(3-42) against Aβ-mediated paralysis. Mammalian neurons were also protected by elevated levels of endogenous STI1 in vitro, and this effect was mainly due to extracellular STI1. Surprisingly, in the 5xFAD mouse model of AD, by overexpressing STI1, we find increased amyloid burden, which amplifies neurotoxicity and worsens spatial memory deficits in these mutants. Increased levels of STI1 disturbed the expression of Aβ-regulating enzymes (BACE1 and MMP-2), suggesting potential mechanisms by which amyloid burden is increased in mice. Notably, we observed that STI1 accumulates in dense-core AD plaques in both 5xFAD mice and human brain tissue. Our findings suggest that elevated levels of STI1 contribute to Aβ accumulation, and that STI1 is deposited in AD plaques in mice and humans. We conclude that despite the protective effects of STI1 in C. elegans and in mammalian cultured neurons, in vivo, the predominant effect of elevated STI1 is deleterious in AD.
Collapse
Affiliation(s)
- Rachel E Lackie
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada
| | - Jose Marques-Lopes
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Valeriy G Ostapchenko
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Sarah Good
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Wing-Yiu Choy
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, Medical Sciences Building, 1151 Richmond St. N, London, N6A 5B7, Canada
| | - Patricija van Oosten-Hawle
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen H Pasternak
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- St. Joseph's Health Care London-Parkwood Institute, St. Joseph's Hospital, 268 Grosvenor St Room A1-015, London, N6A 4V2, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, 1151 Richmond St, London, N6A 3K7, Canada
| | - Vania F Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| |
Collapse
|
23
|
Suk TR, Rousseaux MWC. The role of TDP-43 mislocalization in amyotrophic lateral sclerosis. Mol Neurodegener 2020; 15:45. [PMID: 32799899 PMCID: PMC7429473 DOI: 10.1186/s13024-020-00397-1] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Since its discovery as a primary component in cytoplasmic aggregates in post-mortem tissue of patients with Amyotrophic Lateral Sclerosis (ALS), TAR DNA Binding Protein 43 kDa (TDP-43) has remained a central focus to understand the disease. TDP-43 links both familial and sporadic forms of ALS as mutations are causative for disease and cytoplasmic aggregates are a hallmark of nearly all cases, regardless of TDP-43 mutational status. Research has focused on the formation and consequences of cytosolic protein aggregates as drivers of ALS pathology through both gain- and loss-of-function mechanisms. Not only does aggregation sequester the normal function of TDP-43, but these aggregates also actively block normal cellular processes inevitably leading to cellular demise in a short time span. Although there may be some benefit to therapeutically targeting TDP-43 aggregation, this step may be too late in disease development to have substantial therapeutic benefit. However, TDP-43 pathology appears to be tightly linked with its mislocalization from the nucleus to the cytoplasm, making it difficult to decouple the consequences of nuclear-to-cytoplasmic mislocalization from protein aggregation. Studies focusing on the effects of TDP-43 mislocalization have demonstrated both gain- and loss-of-function consequences including altered splicing regulation, over responsiveness to cellular stressors, increases in DNA damage, and transcriptome-wide changes. Additionally, mutations in TARDBP confer a baseline increase in cytoplasmic TDP-43 thus suggesting that small changes in the subcellular localization of TDP-43 could in fact drive early pathology. In this review, we bring forth the theme of protein mislocalization as a key mechanism underlying ALS, by highlighting the importance of maintaining subcellular proteostasis along with the gain- and loss-of-functional consequences when TDP-43 localization is dysregulated. Additional research, focusing on early events in TDP-43 pathogenesis (i.e. to the protein mislocalization stage) will provide insight into disease mechanisms, therapeutic targets, and novel biomarkers for ALS.
Collapse
Affiliation(s)
- Terry R. Suk
- University of Ottawa Brain and Mind Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Maxime W. C. Rousseaux
- University of Ottawa Brain and Mind Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Eric Poulin Center for Neuromuscular Diseases, Ottawa, Canada
- Ottawa Institute of Systems Biology, Ottawa, Canada
| |
Collapse
|
24
|
Gorantla NV, Das R, Balaraman E, Chinnathambi S. Transition metal nickel prevents Tau aggregation in Alzheimer's disease. Int J Biol Macromol 2020; 156:1359-1365. [DOI: 10.1016/j.ijbiomac.2019.11.176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/24/2019] [Accepted: 11/20/2019] [Indexed: 12/17/2022]
|
25
|
Thiruchittampalam S, Weerasinghe S. Plausible compounds drawn from plants as curative agents for neurodegeneration: An in-silico approach. J Comput Aided Mol Des 2020; 34:1003-1011. [PMID: 32533371 DOI: 10.1007/s10822-020-00322-0] [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: 01/13/2020] [Accepted: 06/07/2020] [Indexed: 11/28/2022]
Abstract
Classification of chemical compounds of plants as a source of medicaments for neurodegenerative diseases through computer screening is an efficient process in drug discovery, in advance of laboratory testing and clinical trials. The onset of neurodegenerative disorders incarcerates both sufferers and their families mentally and financially. This investigation emphasises the search for potent compounds via a computational approach, as an initial path towards the treatment of the neurodegenerative diseases Alzheimer's (AD), Parkinson's (PD), prion, and Huntington's (HD) diseases. The therapeutic strategy considered here is chelation therapy, emanated from the heightened levels of metal ions, which play an imperative role in the pathogenesis of all four neurodegenerative disorders mentioned. Hence, potent compounds from Sri Lankan plants to function as lead compounds have been identified for Cu(II), Fe(III), Zn(II), and Al(III) ions, from a library of around 200 chemical compounds, using an umbrella sampling molecular dynamics computational approach where the chelating ability of compounds for the metal ion is assessed in terms of binding free energy. Calculations reveal that 12 Sri Lankan plants possess compounds that could be considered as starting points of leads for AD, PD and prion disease. However, no compound was potentially useful for the HD category, according to the study. Potential of mean force of Al3+ binding to (-)-5-methylmellin found in Semecarpus walkeri with two representative configurations.
Collapse
|
26
|
Ahmadpour D, Babazadeh R, Nystrom T. Hitchhiking on vesicles: a way to harness age-related proteopathies? FEBS J 2020; 287:5068-5079. [PMID: 32336030 DOI: 10.1111/febs.15345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/06/2020] [Accepted: 04/23/2020] [Indexed: 12/27/2022]
Abstract
Central to proteopathies and leading to most age-related neurodegenerative disorders is a failure in protein quality control (PQC). To harness the toxicity of misfolded and damaged disease proteins, such proteins are either refolded, degraded by temporal PQC, or sequestered by spatial PQC into specific, organelle-associated, compartments within the cell. Here, we discuss the impact of vesicle trafficking pathways in general, and syntaxin 5 in particular, as key players in spatial PQC directing misfolded proteins to the surface of vacuole and mitochondria, which facilitates their clearance and detoxification. Since boosting vesicle trafficking genetically can positively impact on spatial PQC and make cells less sensitive to misfolded disease proteins, we speculate that regulators of such trafficking might serve as therapeutic targets for age-related neurological disorders.
Collapse
Affiliation(s)
- Doryaneh Ahmadpour
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Sweden.,Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - Roja Babazadeh
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Sweden.,Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Thomas Nystrom
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Sweden
| |
Collapse
|
27
|
Gomes LMF, Bataglioli JC, Jussila AJ, Smith JR, Walsby CJ, Storr T. Modification of Aβ Peptide Aggregation via Covalent Binding of a Series of Ru(III) Complexes. Front Chem 2019; 7:838. [PMID: 31921764 PMCID: PMC6915085 DOI: 10.3389/fchem.2019.00838] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, leading to loss of cognition, and eventually death. The disease is characterized by the formation of extracellular aggregates of the amyloid-beta (Aβ) peptide and neurofibrillary tangles of tau protein inside cells, and oxidative stress. In this study, we investigate a series of Ru(III) complexes (Ru-N) derived from NAMI-A in which the imidazole ligand has been substituted for pyridine derivatives, as potential therapeutics for AD. The ability of the Ru-N series to bind to Aβ was evaluated by NMR and ESI-MS, and their influence on the Aβ peptide aggregation process was investigated via electrophoresis gel/western blot, TEM, turbidity, and Bradford assays. The complexes were shown to bind covalently to the Aβ peptide, likely via a His residue. Upon binding, the complexes promote the formation of soluble high molecular weight aggregates, in comparison to peptide precipitation for peptide alone. In addition, TEM analysis supports both amorphous and fibrillar aggregate morphology for Ru-N treatments, while only large amorphous aggregates are observed for peptide alone. Overall, our results show that the Ru-N complexes modulate Aβ peptide aggregation, however, the change in the size of the pyridine ligand does not substantially alter the Aβ aggregation process.
Collapse
Affiliation(s)
- Luiza M F Gomes
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | | | - Allison J Jussila
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Jason R Smith
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Charles J Walsby
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Tim Storr
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
28
|
Ling W, Huang YM, Qiao YC, Zhang XX, Zhao HL. Human Amylin: From Pathology to Physiology and Pharmacology. Curr Protein Pept Sci 2019; 20:944-957. [DOI: 10.2174/1389203720666190328111833] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 12/18/2022]
Abstract
The histopathological hallmark of type 2 diabetes is islet amyloid implicated in the developing treatment options. The major component of human islet amyloid is 37 amino acid peptide known as amylin or islet amyloid polypeptide (IAPP). Amylin is an important hormone that is co-localized, copackaged, and co-secreted with insulin from islet β cells. Physiologically, amylin regulates glucose homeostasis by inhibiting insulin and glucagon secretion. Furthermore, amylin modulates satiety and inhibits gastric emptying via the central nervous system. Normally, human IAPP is soluble and natively unfolded in its monomeric state. Pathologically, human IAPP has a propensity to form oligomers and aggregate. The oligomers show misfolded α-helix conformation and can further convert themselves to β-sheet-rich fibrils as amyloid deposits. The pathological findings and physiological functions of amylin have led to the introduction of pramlintide, an amylin analog, for the treatment of diabetes. The history of amylin’s discovery is a representative example of how a pathological finding can translate into physiological exploration and lead to pharmacological intervention. Understanding the importance of transitioning from pathology to physiology and pharmacology can provide novel insight into diabetes mellitus and Alzheimer's disease.
Collapse
Affiliation(s)
- Wei Ling
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Yan-Mei Huang
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Yong-Chao Qiao
- Department of Laboratory, the Affiliated Hospital of Guilin Medical University, Guilin 541004, China
| | - Xiao-Xi Zhang
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Hai-Lu Zhao
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| |
Collapse
|
29
|
Mrdenovic D, Majewska M, Pieta IS, Bernatowicz P, Nowakowski R, Kutner W, Lipkowski J, Pieta P. Size-Dependent Interaction of Amyloid β Oligomers with Brain Total Lipid Extract Bilayer-Fibrillation Versus Membrane Destruction. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:11940-11949. [PMID: 31328526 DOI: 10.1021/acs.langmuir.9b01645] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Amyloid β, Aβ(1-42), is a component of senile plaques present in the brain of Alzheimer's disease patients and one of the main suspects responsible for pathological consequences of the disease. Herein, we directly visualize the Aβ activity toward a brain-like model membrane and demonstrate that this activity strongly depends on the Aβ oligomer size. PeakForce quantitative nanomechanical mapping mode of atomic force microscopy imaging revealed that the interaction of large-size (LS) Aβ oligomers, corresponding to high-molecular-weight Aβ oligomers, with the brain total lipid extract (BTLE) membrane resulted in accelerated Aβ fibrillogenesis on the membrane surface. Importantly, the fibrillogenesis did not affect integrity of the membrane. In contrast, small-size (SS) Aβ oligomers, corresponding to low-molecular-weight Aβ oligomers, created pores and then disintegrated the BTLE membrane. Both forms of the Aβ oligomers changed nanomechanical properties of the membrane by decreasing its Young's modulus by ∼45%. Our results demonstrated that both forms of Aβ oligomers induce the neurotoxic effect on the brain cells but their action toward the membrane differs significantly.
Collapse
Affiliation(s)
- Dusan Mrdenovic
- Institute of Physical Chemistry , Polish Academy of Sciences , Kasprzaka 44/52 , 01-224 Warsaw , Poland
- Department of Chemistry , University of Guelph , 50 Stone Road East , Guelph , Ontario N1G 2W1 , Canada
| | - Marta Majewska
- Institute of Physical Chemistry , Polish Academy of Sciences , Kasprzaka 44/52 , 01-224 Warsaw , Poland
| | - Izabela S Pieta
- Institute of Physical Chemistry , Polish Academy of Sciences , Kasprzaka 44/52 , 01-224 Warsaw , Poland
| | - Piotr Bernatowicz
- Institute of Physical Chemistry , Polish Academy of Sciences , Kasprzaka 44/52 , 01-224 Warsaw , Poland
| | - Robert Nowakowski
- Institute of Physical Chemistry , Polish Academy of Sciences , Kasprzaka 44/52 , 01-224 Warsaw , Poland
| | - Wlodzimierz Kutner
- Institute of Physical Chemistry , Polish Academy of Sciences , Kasprzaka 44/52 , 01-224 Warsaw , Poland
- Faculty of Mathematics and Natural Sciences, School of Sciences , Cardinal Stefan Wyszynski University in Warsaw , Wóycickiego 1/3 , 01-815 Warsaw , Poland
| | - Jacek Lipkowski
- Department of Chemistry , University of Guelph , 50 Stone Road East , Guelph , Ontario N1G 2W1 , Canada
| | - Piotr Pieta
- Institute of Physical Chemistry , Polish Academy of Sciences , Kasprzaka 44/52 , 01-224 Warsaw , Poland
| |
Collapse
|
30
|
Lee J, Lee JH, Paik SR, Yeom B, Char K. Thermally triggered self-assembly of κ-casein amyloid nanofibrils and their nanomechanical properties. POLYMER 2019. [DOI: 10.1016/j.polymer.2019.121626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
31
|
Foley AR, Finn TS, Kung T, Hatami A, Lee HW, Jia M, Rolandi M, Raskatov JA. Trapping and Characterization of Nontoxic Aβ42 Aggregation Intermediates. ACS Chem Neurosci 2019; 10:3880-3887. [PMID: 31319029 DOI: 10.1021/acschemneuro.9b00340] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Amyloid β (Aβ) 42 is an aggregation-prone peptide and the believed seminal etiological agent of Alzheimer's disease (AD). Intermediates of Aβ42 aggregation, commonly referred to as diffusible oligomers, are considered to be among the most toxic forms of the peptide. Here, we studied the effect of the age-related epimerization of Ser26 (i.e., S26s chiral edit) in Aβ42 and discovered that this subtle molecular change led to reduced fibril formation propensity. Surprisingly, the resultant soluble aggregates were nontoxic. To gain insight into the structural changes that occurred in the peptide upon S26s substitution, the system was probed using an array of biophysical and biochemical methods. These experiments consistently pointed to the stabilization of aggregation intermediates in the Aβ42-S26s system. To better understand the changes arising as a consequence of the S26s substitution, molecular level structural studies were performed. Using a combined nuclear magnetic resonance (NMR)- and density functional theory (DFT)-computational approach, we found that the S26s chiral edit induced only local structural changes in the Gly25-Ser26-Asn27 region. Interestingly, these subtle changes enabled the formation of an intramolecular Ser26-Asn27 H-bond, which disrupted the ability of Asn27 to engage in the fibrillogenic side chain-to-side chain H-bonding pattern. This reveals that intermolecular stabilizing interactions between Asn27 side chains are a key element controlling Aβ42 aggregation and toxicity.
Collapse
Affiliation(s)
- Alejandro R. Foley
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California 95064, United States
| | - Thomas S. Finn
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California 95064, United States
| | - Timothy Kung
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California 95064, United States
| | - Asa Hatami
- Sangamo Therapeutics, Richmond, California 94804, United States
| | - Hsiau-Wei Lee
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California 95064, United States
| | - Manping Jia
- Department of Electrical Engineering, University of California Santa Cruz, Santa Cruz, California 95064, United States
| | - Marco Rolandi
- Department of Electrical Engineering, University of California Santa Cruz, Santa Cruz, California 95064, United States
| | - Jevgenij A. Raskatov
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California 95064, United States
| |
Collapse
|
32
|
Penke B, Bogár F, Paragi G, Gera J, Fülöp L. Key Peptides and Proteins in Alzheimer's Disease. Curr Protein Pept Sci 2019; 20:577-599. [PMID: 30605056 DOI: 10.2174/1389203720666190103123434] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/03/2018] [Accepted: 12/27/2018] [Indexed: 02/02/2023]
Abstract
Alzheimer's Disease (AD) is a form of progressive dementia involving cognitive impairment, loss of learning and memory. Different proteins (such as amyloid precursor protein (APP), β- amyloid (Aβ) and tau protein) play a key role in the initiation and progression of AD. We review the role of the most important proteins and peptides in AD pathogenesis. The structure, biosynthesis and physiological role of APP are shortly summarized. The details of trafficking and processing of APP to Aβ, the cytosolic intracellular Aβ domain (AICD) and small soluble proteins are shown, together with other amyloid-forming proteins such as tau and α-synuclein (α-syn). Hypothetic physiological functions of Aβ are summarized. The mechanism of conformational change, the formation and the role of neurotoxic amyloid oligomeric (oAβ) are shown. The fibril formation process and the co-existence of different steric structures (U-shaped and S-shaped) of Aβ monomers in mature fibrils are demonstrated. We summarize the known pathogenic and non-pathogenic mutations and show the toxic interactions of Aβ species after binding to cellular receptors. Tau phosphorylation, fibrillation, the molecular structure of tau filaments and their toxic effect on microtubules are shown. Development of Aβ and tau imaging in AD brain and CSF as well as blood biomarkers is shortly summarized. The most probable pathomechanisms of AD including the toxic effects of oAβ and tau; the three (biochemical, cellular and clinical) phases of AD are shown. Finally, the last section summarizes the present state of Aβ- and tau-directed therapies and future directions of AD research and drug development.
Collapse
Affiliation(s)
- Botond Penke
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary
| | - Ferenc Bogár
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary.,MTA-SZTE Biomimetic Systems Research Group, University of Szeged, H-6720 Szeged, Dom square 8, Hungary
| | - Gábor Paragi
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, H-6720 Szeged, Dom square 8, Hungary.,Institute of Physics, University of Pécs, H-7624 Pecs, Ifjusag utja 6, Hungary
| | - János Gera
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary
| | - Lívia Fülöp
- Department of Medical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom square 8, Szeged, H-6720, Hungary
| |
Collapse
|
33
|
Robison LS, Popescu DL, Anderson ME, Francis N, Hatfield J, Sullivan JK, Beigelman SI, Xu F, Anderson BJ, Van Nostrand WE, Robinson JK. Long-term voluntary wheel running does not alter vascular amyloid burden but reduces neuroinflammation in the Tg-SwDI mouse model of cerebral amyloid angiopathy. J Neuroinflammation 2019; 16:144. [PMID: 31296239 PMCID: PMC6621983 DOI: 10.1186/s12974-019-1534-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cardiovascular exercise (CVE) has been shown to be protective against cognitive decline in aging and the risk for dementias, including Alzheimer's Disease (AD). CVE has also been shown to have several beneficial effects on brain pathology and behavioral impairments in mouse models of AD; however, no studies have specifically examined the effects of CVE on cerebral amyloid angiopathy (CAA), which is the accumulation of amyloid-beta (Aβ) in the cerebral vasculature. CAA may be uniquely susceptible to beneficial effects of CVE interventions due to the location and nature of the pathology. Alternatively, CVE may exacerbate CAA pathology, due to added stress on already compromised cerebral vasculature. METHODS In the current study, we examined the effects of CVE over many months in mice, thereby modeling a lifelong commitment to CVE in humans. We assessed this voluntary CVE in Tg-SwDI mice, a transgenic mouse model of CAA that exhibits behavioral deficits, fibrillar vascular Aβ pathology, and significant perivascular neuroinflammation. Various "doses" of exercise intervention (0 h ("Sedentary"), 1 h, 3 h, 12 h access to running wheel) were assessed from ~ 4 to 12 months of age for effects on physiology, behavior/cognitive performance, and pathology. RESULTS The 12 h group performed the greatest volume of exercise, whereas the 1 h and 3 h groups showed high levels of exercise intensity, as defined by more frequent and longer duration running bouts. Tg-SwDI mice exhibited significant cerebral vascular Aβ pathology and increased expression of pro-inflammatory cytokines as compared to WT controls. Tg-SwDI mice did not show motor dysfunction or altered levels of anxiety or sociability compared to WT controls, though Tg-SwDI animals did appear to exhibit a reduced tendency to explore novel environments. At all running levels, CAA pathology in Tg-SwDI mice was not significantly altered, but 12-h high-volume exercise showed increased insoluble Aβ burden. However, CVE attenuated the expression of pro-inflammatory cytokines TNF-α and IL-6 and was generally effective at enhancing motor function and reducing anxiety-like behavior in Tg-SwDI mice, though alterations in learning and memory tasks were varied. CONCLUSIONS Taken together, these results suggest that CAA can still develop regardless of a lifespan of substantial CVE, although downstream effects on neuroinflammation may be reduced and functional outcomes improved.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Psychology, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA.,Present Address: Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA
| | - Dominique L Popescu
- Department of Psychology, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA.,Present Address: George and Anne Ryan Institute for Neuroscience and Department of Psychology, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
| | - Maria E Anderson
- Department of Psychology, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA.,Present Address: Department of Psychology, Farmingdale State University, 2350 Broadhollow Rd, Farmingdale, NY, 11735, USA
| | - Nikita Francis
- Department of Psychology, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA.,Present Address: George and Anne Ryan Institute for Neuroscience and Department of Psychology, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
| | - Joshua Hatfield
- George & Anne Ryan Institute for Neuroscience and Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
| | - Joseph K Sullivan
- Present Address: New York Medical College, School of Medicine, 40 Sunshine Cottage Rd, Valhalla, NY, 10595, USA
| | - Steven I Beigelman
- Department of Psychology, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA
| | - Feng Xu
- George & Anne Ryan Institute for Neuroscience and Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
| | - Brenda J Anderson
- Department of Psychology, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA
| | - William E Van Nostrand
- George & Anne Ryan Institute for Neuroscience and Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
| | - John K Robinson
- Department of Psychology, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA. .,Present Address: George and Anne Ryan Institute for Neuroscience and Department of Psychology, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA.
| |
Collapse
|
34
|
Das S, Pahari S, Sarmah S, Rohman MA, Paul D, Jana M, Singha Roy A. Lysozyme-luteolin binding: molecular insights into the complexation process and the inhibitory effects of luteolin towards protein modification. Phys Chem Chem Phys 2019; 21:12649-12666. [PMID: 31157335 DOI: 10.1039/c9cp01128e] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the proposed work, the complexation of bioactive flavonoid luteolin with hen egg white lysozyme (HEWL) along with its inhibitory influence on HEWL modification has been explored with the help of multi-spectroscopic and computational methods. The binding affinity has been observed to be moderate in nature (in the order of 104 M-1) and the static quenching mechanism was found to be involved in the fluorescence quenching process. The binding constant (Kb) shows a progressive increase with the increase in temperature from (4.075 ± 0.046 × 104 M-1) at 293 K to (6.962 ± 0.024 × 104 M-1) at 313 K under experimental conditions. Spectroscopic measurements along with molecular docking calculations suggest that Trp62 is involved in the binding site of luteolin within the geometry of HEWL. The positive changes in enthalpy (ΔH = +19.99 ± 0.65 kJ mol-1) as well as entropy (ΔS = +156.28 ± 2.00 J K-1 mol-1) are indicative of the presence of hydrophobic forces that stabilize the HEWL-luteolin complex. The micro-environment around the Trp residues showed an increase in hydrophobicity as indicated by synchronous fluorescence (SFS), three dimensional fluorescence (3D) and red edge excitation (REES) studies. The % α-helix of HEWL showed a marked reduction upon binding with luteolin as indicated by circular dichroism (CD) and Fourier-transform infrared spectroscopy (FTIR) studies. Moreover, luteolin is situated at a distance of 4.275 ± 0.004 nm from the binding site as indicated by FRET theory, and the rate of energy transfer kET (0.063 ± 0.004 ns-1) has been observed to be faster than the donor decay rate (1/τD = 0.606 ns-1), which is indicative of the non-radiative energy transfer during complexation. Leaving aside the binding study, luteolin showed promising inhibitory effects towards the d-ribose mediated glycation of HEWL as well as towards HEWL fibrillation as studied by fluorescence emission and imaging studies. Excellent correlation with the experimental observations as well as precise location and dynamics of luteolin within the binding site has been obtained from molecular docking and molecular dynamics simulation studies.
Collapse
Affiliation(s)
- Sourav Das
- Department of Chemistry, National Institute of Technology, Shillong 793003, Meghalaya, India.
| | - Somdev Pahari
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela 769008, India.
| | - Sharat Sarmah
- Department of Chemistry, National Institute of Technology, Shillong 793003, Meghalaya, India.
| | - Mostofa Ataur Rohman
- Centre for Advanced Studies, Department of Chemistry, North-Eastern Hill University, Shillong 793022, India
| | - Debojit Paul
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Madhurima Jana
- Molecular Simulation Laboratory, Department of Chemistry, National Institute of Technology, Rourkela 769008, India.
| | - Atanu Singha Roy
- Department of Chemistry, National Institute of Technology, Shillong 793003, Meghalaya, India.
| |
Collapse
|
35
|
Mehrazma B, Rauk A. Exploring Amyloid-β Dimer Structure Using Molecular Dynamics Simulations. J Phys Chem A 2019; 123:4658-4670. [PMID: 31082235 DOI: 10.1021/acs.jpca.8b11251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A major hallmark of Alzheimer's disease (AD) is the aggregation of amyloid-β peptides in the brains of people afflicted by the disease. The exact pathway to this catastrophic event is unknown. In this work, a total of 9.5 μs molecular dynamics simulations have been performed to investigate the structure and dynamics of the smallest form of toxic Aβ oligomers, i.e., the Aβ dimers. This study suggests that specific hydrophobic regions are vital in the aggregation process. Different possible structures for Aβ dimers are reported along with their relative binding affinity. These data may be used to design better Aβ-aggregation inhibitors. The diversity of the dimer structures suggests several aggregation pathways.
Collapse
Affiliation(s)
- Banafsheh Mehrazma
- Department of Chemistry , University of Calgary , Calgary AB , Canada T2N 1N4
| | - Arvi Rauk
- Department of Chemistry , University of Calgary , Calgary AB , Canada T2N 1N4
| |
Collapse
|
36
|
Marciani DJ. Promising Results from Alzheimer's Disease Passive Immunotherapy Support the Development of a Preventive Vaccine. RESEARCH 2019; 2019:5341375. [PMID: 31549066 PMCID: PMC6750119 DOI: 10.34133/2019/5341375] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/18/2019] [Indexed: 12/23/2022]
Abstract
The apparently near-term effects of the monoclonal antibody BAN2401 in slowing the progression of prodromal Alzheimer's disease (AD) has created cautious optimism about the therapeutic use of antibodies that neutralize cytotoxic soluble amyloid-β aggregates, rather than removing plaque. Plaque being protective, as it immobilizes cytotoxic amyloid-β, rather than AD's causative agent. The presence of natural antibodies against cytotoxic amyloid-β implies the existence of a protective anti-AD immunity. Hence, for vaccines to induce a similar immunoresponse that prevents and/or delays the onset of AD, they must have adjuvants that stimulate a sole anti-inflammatory Th2 immunity, plus immunogens that induce a protective immunoresponse against diverse cytotoxic amyloid-β conformers. Indeed, amyloid-β pleomorphism may explain the lack of long-term protection by monoclonal antibodies that neutralize single conformers, like aducanumab. A situation that would allow new cytotoxic conformers to escape neutralization by previously effective monoclonal antibodies. Stimulation of a vaccine's effective immunoresponse would require the concurrent delivery of immunogen to dendritic cells and their priming, to induce a polarized Th2 immunity. An immunoresponse that would produce besides neutralizing antibodies against neurotoxic amyloid-β oligomers, anti-inflammatory cytokines; preventing inflammation that aggravates AD. Because of age-linked immune decline, vaccines would be significantly more effective in preventing, rather than treating AD. Considering the amyloid-β's role in tau's pathological hyperphosphorylation and their synergism in AD, the development of preventive vaccines against both amyloid-β and tau should be considered. Due to convenience and cost, vaccines may be the only option available to many countries to forestall the impending AD epidemic.
Collapse
Affiliation(s)
- D J Marciani
- Qantu Therapeutics, Inc., 612 E. Main Street, Lewisville, TX 75057, USA
| |
Collapse
|
37
|
Paul A, Zhang BD, Mohapatra S, Li G, Li YM, Gazit E, Segal D. Novel Mannitol-Based Small Molecules for Inhibiting Aggregation of α-Synuclein Amyloids in Parkinson's Disease. Front Mol Biosci 2019; 6:16. [PMID: 30968030 PMCID: PMC6438916 DOI: 10.3389/fmolb.2019.00016] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/01/2019] [Indexed: 11/16/2022] Open
Abstract
The aggregation of the amyloidogenic protein α-synuclein (α-Syn) into toxic oligomers and mature fibrils is the major pathological hallmark of Parkinson's disease (PD). Small molecules that inhibit α-Syn aggregation thus may be useful therapeutics for PD. Mannitol and naphthoquinone-tryptophan (NQTrp) have been shown in the past to inhibit α-Syn aggregation by different mechanisms. Herein, we tested whether the conjugation of Mannitol and NQTrp may result in enhance efficacy toward α-Syn. The molecules were conjugated either by a click linker or via a PEG linker. The effect of the conjugate molecules on α-Syn aggregation in vitro was monitored using Thioflavin T fluorescence assay, circular dichroism, transmission electron microscopy, and Congo red birefringence assay. One of the conjugate molecules was found to be more effective than the two parent molecules and as effective as a mixture of the two. The conjugate molecules attenuated the disruptive effect of α-Syn on artificial membrane of Large Unilamellar Vesicles as monitored by dye leakage assay. The conjugates were found to be have low cytotoxicity and reduced toxicity of α-Syn toward SH-SY5Y neuroblastoma cells. These novel designed entities can be attractive scaffold for the development of therapeutic agents for PD.
Collapse
Affiliation(s)
- Ashim Paul
- School of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Bo-Dou Zhang
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Satabdee Mohapatra
- School of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Gao Li
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Yan-Mei Li
- Department of Chemistry, Tsinghua University, Beijing, China.,Institute of Parkinson Disease, Beijing Institute for Brain Disorders, Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Ehud Gazit
- School of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Segal
- School of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel.,Sagol Interdisciplinary School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
38
|
Aducanumab Therapy Ameliorates Calcium Overload in a Mouse Model of Alzheimer's Disease. J Neurosci 2018; 37:4430-4432. [PMID: 28446659 DOI: 10.1523/jneurosci.0420-17.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 01/18/2023] Open
|
39
|
NEDDylation promotes nuclear protein aggregation and protects the Ubiquitin Proteasome System upon proteotoxic stress. Nat Commun 2018; 9:4376. [PMID: 30349034 PMCID: PMC6197266 DOI: 10.1038/s41467-018-06365-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 08/24/2018] [Indexed: 11/23/2022] Open
Abstract
Spatial management of stress-induced protein aggregation is an integral part of the proteostasis network. Protein modification by the ubiquitin-like molecule NEDD8 increases upon proteotoxic stress and it is characterised by the formation of hybrid NEDD8/ubiquitin conjugates. However, the biological significance of this response is unclear. Combination of quantitative proteomics with biological analysis shows that, during proteotoxic stress, NEDDylation promotes nuclear protein aggregation, including ribosomal proteins as a major group. This correlates with protection of the nuclear Ubiquitin Proteasome System from stress-induced dysfunction. Correspondingly, we show that NEDD8 compromises ubiquitination and prevents targeting and processing of substrates by the proteasome. Moreover, we identify HUWE1 as a key E3-ligase that is specifically required for NEDDylation during proteotoxic stress. The study reveals a specific role for NEDD8 in nuclear protein aggregation upon stress and is consistent with the concept that transient aggregate formation is part of a defence mechanism against proteotoxicity. Protein NEDDylation increases upon proteotoxic stress but the function of this response remains to be elucidated. Here, the authors show that NEDDylation contributes to the cellular defence against proteotoxicity by promoting nuclear protein aggregation and protecting the ubiquitin proteasome system.
Collapse
|
40
|
Chen JY, Parekh M, Seliman H, Bakshinskaya D, Dai W, Kwan K, Chen KY, Liu AYC. Heat shock promotes inclusion body formation of mutant huntingtin (mHtt) and alleviates mHtt-induced transcription factor dysfunction. J Biol Chem 2018; 293:15581-15593. [PMID: 30143534 PMCID: PMC6177601 DOI: 10.1074/jbc.ra118.002933] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 08/22/2018] [Indexed: 01/08/2023] Open
Abstract
PolyQ-expanded huntingtin (mHtt) variants form aggregates, termed inclusion bodies (IBs), in individuals with and models of Huntington's disease (HD). The role of IB versus diffusible mHtt in neurotoxicity remains unclear. Using a ponasterone (PA)-inducible cell model of HD, here we evaluated the effects of heat shock on the appearance and functional outcome of Htt103QExon1-EGFP expression. Quantitative image analysis indicated that 80-90% of this mHtt protein initially appears as "diffuse" signals in the cytosol, with IBs forming at high mHtt expression. A 2-h heat shock during the PA induction reduced the diffuse signal, but greatly increased mHtt IB formation in both cytosol and nucleus. Dose- and time-dependent mHtt expression suggested that nucleated polymerization drives IB formation. RNA-mediated knockdown of heat shock protein 70 (HSP70) and heat shock cognate 70 protein (HSC70) provided evidence for their involvement in promoting diffuse mHtt to form IBs. Reporter gene assays assessing the impacts of diffuse versus IB mHtt showed concordance of diffuse mHtt expression with the repression of heat shock factor 1, cAMP-responsive element-binding protein (CREB), and NF-κB activity. CREB repression was reversed by heat shock coinciding with mHtt IB formation. In an embryonic striatal neuron-derived HD model, the chemical chaperone sorbitol similarly promoted the structuring of diffuse mHtt into IBs and supported cell survival under stress. Our results provide evidence that mHtt IB formation is a chaperone-supported cellular coping mechanism that depletes diffusible mHtt conformers, alleviates transcription factor dysfunction, and promotes neuron survival.
Collapse
Affiliation(s)
- Justin Y Chen
- From the Department of Cell Biology and Neuroscience and
| | - Miloni Parekh
- From the Department of Cell Biology and Neuroscience and
| | - Hadear Seliman
- From the Department of Cell Biology and Neuroscience and
| | | | - Wei Dai
- From the Department of Cell Biology and Neuroscience and
| | - Kelvin Kwan
- From the Department of Cell Biology and Neuroscience and
| | - Kuang Yu Chen
- Department of Chemistry and Chemical Biology, Rutgers State University of New Jersey, Piscataway, New Jersey 08854
| | - Alice Y C Liu
- From the Department of Cell Biology and Neuroscience and
| |
Collapse
|
41
|
Abstract
The cellular prion protein, PrPC, is a small, cell surface glycoprotein with a function that is currently somewhat ill defined. It is also the key molecule involved in the family of neurodegenerative disorders called transmissible spongiform encephalopathies, which are also known as prion diseases. The misfolding of PrPC to a conformationally altered isoform, designated PrPTSE, is the main molecular process involved in pathogenesis and appears to precede many other pathologic and clinical manifestations of disease, including neuronal loss, astrogliosis, and cognitive loss. PrPTSE is also believed to be the major component of the infectious "prion," the agent responsible for disease transmission, and preparations of this protein can cause prion disease when inoculated into a naïve host. Thus, understanding the biochemical and biophysical properties of both PrPC and PrPTSE, and ultimately the mechanisms of their interconversion, is critical if we are to understand prion disease biology. Although entire books could be devoted to research pertaining to the protein, herein we briefly review the state of knowledge of prion biochemistry, including consideration of prion protein structure, function, misfolding, and dysfunction.
Collapse
Affiliation(s)
- Andrew C Gill
- School of Chemistry, Joseph Banks Laboratories, University of Lincoln, Lincoln, United Kingdom; Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| | - Andrew R Castle
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
42
|
Mehrazma B, Opare S, Petoyan A, Rauk A. d-Amino Acid Pseudopeptides as Potential Amyloid-Beta Aggregation Inhibitors. Molecules 2018; 23:E2387. [PMID: 30231520 PMCID: PMC6225248 DOI: 10.3390/molecules23092387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/06/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022] Open
Abstract
A causative factor for neurotoxicity associated with Alzheimer's disease is the aggregation of the amyloid-β (Aβ) peptide into soluble oligomers. Two all d-amino acid pseudo-peptides, SGB1 and SGD1, were designed to stop the aggregation. Molecular dynamics (MD) simulations have been carried out to study the interaction of the pseudo-peptides with both Aβ13⁻23 (the core recognition site of Aβ) and full-length Aβ1⁻42. Umbrella sampling MD calculations have been used to estimate the free energy of binding, ∆G, of these peptides to Aβ13⁻23. The highest ∆Gbinding is found for SGB1. Each of the pseudo-peptides was also docked to Aβ1⁻42 and subjected up to seven microseconds of all atom molecular dynamics simulations. The resulting structures lend insight into how the dynamics of Aβ1⁻42 are altered by complexation with the pseudo-peptides and confirmed that SGB1 may be a better candidate for developing into a drug to prevent Alzheimer's disease.
Collapse
Affiliation(s)
- Banafsheh Mehrazma
- Department of Chemistry, University of Calgary; Calgary, AB T2N 1N4, Canada.
| | - Stanley Opare
- Department of Chemistry, University of Calgary; Calgary, AB T2N 1N4, Canada.
| | - Anahit Petoyan
- Department of Chemistry, University of Calgary; Calgary, AB T2N 1N4, Canada.
| | - Arvi Rauk
- Department of Chemistry, University of Calgary; Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
43
|
Mechanism of aggregation and membrane interactions of mammalian prion protein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018. [DOI: 10.1016/j.bbamem.2018.02.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
44
|
Katyal N, Agarwal M, Sen R, Kumar V, Deep S. Paradoxical Effect of Trehalose on the Aggregation of α-Synuclein: Expedites Onset of Aggregation yet Reduces Fibril Load. ACS Chem Neurosci 2018; 9:1477-1491. [PMID: 29601727 DOI: 10.1021/acschemneuro.8b00056] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Aggregation of α-synuclein is closely connected to the pathology of Parkinson's disease. The phenomenon involves multiple steps, commenced by partial misfolding and eventually leading to mature amyloid fibril formation. Trehalose, a widely accepted osmolyte, has been shown previously to inhibit aggregation of various globular proteins owing to its ability to prevent the initial unfolding of protein. In this study, we have examined if it behaves in a similar fashion with intrinsically disordered protein α-synuclein and possesses the potential to act as therapeutic agent against Parkinson's disease. It was observed experimentally that samples coincubated with trehalose fibrillate faster compared to the case in its absence. Molecular dynamics simulations suggested that this initial acceleration is manifestation of trehalose's tendency to perturb the conformational transitions between different conformers of monomeric protein. It stabilizes the aggregation prone "extended" conformer of α-synuclein, by binding to its exposed acidic residues of the C terminus. It also favors the β-rich oligomers once formed. Interestingly, the total fibrils formed are still promisingly less since it accelerates the competing pathway toward formation of amorphous aggregates.
Collapse
Affiliation(s)
- Nidhi Katyal
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Manish Agarwal
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Raktim Sen
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Vinay Kumar
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| |
Collapse
|
45
|
Specification of Physiologic and Disease States by Distinct Proteins and Protein Conformations. Cell 2017; 171:1001-1014. [PMID: 29149602 DOI: 10.1016/j.cell.2017.10.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022]
Abstract
Protein conformational states-from intrinsically disordered ensembles to amyloids that underlie the self-templating, infectious properties of prion-like proteins-have attracted much attention. Here, we highlight the diversity, including differences in biophysical properties, that drive distinct biological functions and pathologies among self-templating proteins. Advances in chemical genomics, gene editing, and model systems now permit deconstruction of the complex interplay between these protein states and the host factors that react to them. These methods reveal that conformational switches modulate normal and abnormal information transfer and that intimate relationships exist between the intrinsic function of proteins and the deleterious consequences of their misfolding.
Collapse
|
46
|
Asymmetric Inheritance of Aggregated Proteins and Age Reset in Yeast Are Regulated by Vac17-Dependent Vacuolar Functions. Cell Rep 2017; 16:826-38. [PMID: 27373154 PMCID: PMC4963537 DOI: 10.1016/j.celrep.2016.06.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/29/2016] [Accepted: 05/31/2016] [Indexed: 11/24/2022] Open
Abstract
Age can be reset during mitosis in both yeast and stem cells to generate a young daughter cell from an aged and deteriorated one. This phenomenon requires asymmetry-generating genes (AGGs) that govern the asymmetrical inheritance of aggregated proteins. Using a genome-wide imaging screen to identify AGGs in Saccharomyces cerevisiae, we discovered a previously unknown role for endocytosis, vacuole fusion, and the myosin-dependent adaptor protein Vac17 in asymmetrical inheritance of misfolded proteins. Overproduction of Vac17 increases deposition of aggregates into cytoprotective vacuole-associated sites, counteracts age-related breakdown of endocytosis and vacuole integrity, and extends replicative lifespan. The link between damage asymmetry and vesicle trafficking can be explained by a direct interaction between aggregates and vesicles. We also show that the protein disaggregase Hsp104 interacts physically with endocytic vesicle-associated proteins, such as the dynamin-like protein, Vps1, which was also shown to be required for Vac17-dependent sequestration of protein aggregates. These data demonstrate that two physiognomies of aging-reduced endocytosis and protein aggregation-are interconnected and regulated by Vac17.
Collapse
|
47
|
Kurochkin IV, Guarnera E, Berezovsky IN. Insulin-Degrading Enzyme in the Fight against Alzheimer's Disease. Trends Pharmacol Sci 2017; 39:49-58. [PMID: 29132916 DOI: 10.1016/j.tips.2017.10.008] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/18/2017] [Accepted: 10/23/2017] [Indexed: 11/19/2022]
Abstract
After decades of research and clinical trials there is still no cure for Alzheimer's disease (AD). While impaired clearance of amyloid beta (Aβ) peptides is considered as one of the major causes of AD, it was recently complemented by a potential role of other toxic amyloidogenic species. Insulin-degrading enzyme (IDE) is the proteolytic culprit of various β-forming peptides, both extracellular and intracellular. On the basis of demonstrated allosteric activation of IDE against Aβ, it is possible to propose a new strategy for the targeted IDE-based cleansing of different toxic aggregation-prone peptides. Consequently, specific allosteric activation of IDE coupled with state-of-the-art compound delivery and CRISP-Cas9 technique of transgene insertion can be instrumental in the fight against AD and related neurodegenerative maladies.
Collapse
Affiliation(s)
- Igor V Kurochkin
- Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-ku, Kobe 651-2271, Japan
| | - Enrico Guarnera
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, 07-01, Matrix, Singapore 138671
| | - Igor N Berezovsky
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, 07-01, Matrix, Singapore 138671; Department of Biological Sciences (DBS), National University of Singapore (NUS), 8 Medical Drive, Singapore 117579.
| |
Collapse
|
48
|
Lee J, Lee JH, Yeom B, Char K. Layer-by-Layer Assembly of κ-Casein Amyloid Fibrils for the Preparation of Hollow Microcapsules. MACROMOL CHEM PHYS 2017. [DOI: 10.1002/macp.201700382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Jubong Lee
- The National Creative Research Initiative Center for Intelligent Hybrids; The WCU Program of Chemical Convergence for Energy & Environment; School of Chemical & Biological Engineering; Seoul National University; Seoul 08826 Korea
| | - Ji-Hye Lee
- The National Creative Research Initiative Center for Intelligent Hybrids; The WCU Program of Chemical Convergence for Energy & Environment; School of Chemical & Biological Engineering; Seoul National University; Seoul 08826 Korea
| | - Bongjun Yeom
- Department of Chemical Engineering; Myongji University; Yongin 17058 Korea
| | - Kookheon Char
- The National Creative Research Initiative Center for Intelligent Hybrids; The WCU Program of Chemical Convergence for Energy & Environment; School of Chemical & Biological Engineering; Seoul National University; Seoul 08826 Korea
| |
Collapse
|
49
|
Polyglutamine expansion diseases: More than simple repeats. J Struct Biol 2017; 201:139-154. [PMID: 28928079 DOI: 10.1016/j.jsb.2017.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/24/2017] [Accepted: 09/15/2017] [Indexed: 12/27/2022]
Abstract
Polyglutamine (polyQ) repeat-containing proteins are widespread in the human proteome but only nine of them are associated with highly incapacitating neurodegenerative disorders. The genetic expansion of the polyQ tract in disease-related proteins triggers a series of events resulting in neurodegeneration. The polyQ tract plays the leading role in the aggregation mechanism, but other elements modulate the aggregation propensity in the context of the full-length proteins, as implied by variations in the length of the polyQ tract required to trigger the onset of a given polyQ disease. Intrinsic features such as the presence of aggregation-prone regions (APRs) outside the polyQ segments and polyQ-flanking sequences, which synergistically participate in the aggregation process, are emerging for several disease-related proteins. The inherent polymorphic structure of polyQ stretches places the polyQ proteins in a central position in protein-protein interaction networks, where interacting partners may additionally shield APRs or reshape the aggregation course. Expansion of the polyQ tract perturbs the cellular homeostasis and contributes to neuronal failure by modulating protein-protein interactions and enhancing toxic oligomerization. Post-translational modifications further regulate self-assembly either by directly altering the intrinsic aggregation propensity of polyQ proteins, by modulating their interaction with different macromolecules or by modifying their withdrawal by the cell quality control machinery. Here we review the recent data on the multifaceted aggregation pathways of disease-related polyQ proteins, focusing on ataxin-3, the protein mutated in Machado-Joseph disease. Further mechanistic understanding of this network of events is crucial for the development of effective therapies for polyQ diseases.
Collapse
|
50
|
Brain ureido degenerative protein modifications are associated with neuroinflammation and proteinopathy in Alzheimer's disease with cerebrovascular disease. J Neuroinflammation 2017; 14:175. [PMID: 28865468 PMCID: PMC5581431 DOI: 10.1186/s12974-017-0946-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/23/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Brain degenerative protein modifications (DPMs) are associated with the apparition and progression of dementia, and at the same time, Alzheimer's disease with cerebrovascular disease (AD + CVD) is the most prevalent form of dementia in the elder population. Thus, understanding the role(s) of brain DPMs in this dementia subtype may provide novel insight on the disease pathogenesis and may aid on the development of novel diagnostic and therapeutic tools. Two essential DPMs known to promote inflammation in several human diseases are the ureido DPMs (uDPMs) arginine citrullination and lysine carbamylation, although they have distinct enzymatic and non-enzymatic origins, respectively. Nevertheless, the implication of uDPMs in the neuropathology of dementia remains poorly understood. METHODS In this study, we use the state-of-the-art, ultracentrifugation-electrostatic repulsion hydrophilic interaction chromatography (UC-ERLIC)-coupled mass spectrometry technology to undertake a comparative characterization of uDPMs in the soluble and particulate postmortem brain fractions of subjects diagnosed with AD + CVD and age-matched controls. RESULTS An increase in the formation of uDPMs was observed in all the profiled AD + CVD brains. Citrulline-containing proteins were found more abundant in the soluble fraction of AD + CVD whereas homocitrulline-containing proteins were preferentially abundant in the particulate fraction of AD + CVD brains. Several dementia-specific citrulline residues were also identified in soluble proteins previously categorized as pro-immunogenic, which include the receptor P2X7, alpha-internexin, GFAP, CNP, MBP, and histones. Similarly, diverse dementia-specific homocitrulline residues were also observed in the particulate fractions of AD + CVD in proteins that have been vastly implicated in neuropathology. Intriguingly, we also found that the amino acids immediately flanking arginine residues may specifically influence the increase in protein citrullination. CONCLUSIONS Taken together, these results indicate that uDPMs widely contribute to the pathophysiology of AD + CVD by promoting neuroinflammation and proteinopathy. Furthermore, the obtained results could help to identify disease-associated proteins that can act as potential targets for therapeutic intervention or as novel biomarkers of specific neuropathology.
Collapse
|