1
|
Basha S, Mukunda DC, Pai AR, Mahato KK. Assessing amyloid fibrils and amorphous aggregates: A review. Int J Biol Macromol 2025; 311:143725. [PMID: 40324497 DOI: 10.1016/j.ijbiomac.2025.143725] [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: 01/23/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Protein misfolding and aggregation play a central role in the progression of neurodegenerative diseases such as Alzheimer's and Parkinson's. These aggregates manifest either as structured amyloid fibrils enriched in β-sheet conformations or as irregular amorphous aggregates with diverse morphologies. Understanding their formation, structure, and behavior is critical for deciphering disease mechanisms and developing targeted diagnostics and therapeutics. This review presents an integrated overview of both conventional and advanced techniques used to detect, distinguish, and structurally characterize these protein aggregates. It covers a range of spectroscopic and spectrometric tools, such as fluorescence, Raman, and mass spectrometry that facilitate aggregate identification. Microscopy methods, including atomic force and electron microscopy, are highlighted for morphological analysis. The review also discusses in situ detection strategies using fluorescent dyes, conformation-specific antibodies, enzymatic reporters, and real-time imaging. Separation methods like centrifugation, electrophoresis, and chromatography are outlined alongside structural analysis tools such as X-ray diffraction. Furthermore, the growing utility of computational approaches and artificial intelligence in predicting aggregation propensities and integrating biological data is emphasized. By critically evaluating each method's capabilities and limitations, this review provides a practical and forward-looking resource for researchers studying the complex landscape of protein aggregation.
Collapse
Affiliation(s)
- Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | | | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
2
|
Guo Z, Chiesa G, Yin J, Sanford A, Meier S, Khalil AS, Cheng JX. Structural Mapping of Protein Aggregates in Live Cells Modeling Huntington's Disease. Angew Chem Int Ed Engl 2024; 63:e202408163. [PMID: 38880765 PMCID: PMC11781839 DOI: 10.1002/anie.202408163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
While protein aggregation is a hallmark of many neurodegenerative diseases, acquiring structural information on protein aggregates inside live cells remains challenging. Traditional microscopy does not provide structural information on protein systems. Routinely used fluorescent protein tags, such as Green Fluorescent Protein (GFP), might perturb native structures. Here, we report a counter-propagating mid-infrared photothermal imaging approach enabling mapping of secondary structure of protein aggregates in live cells modeling Huntington's disease. By comparing mid-infrared photothermal spectra of label-free and GFP-tagged huntingtin inclusions, we demonstrate that GFP fusions indeed perturb the secondary structure of aggregates. By implementing spectra with small spatial step for dissecting spectral features within sub-micrometer distances, we reveal that huntingtin inclusions partition into a β-sheet-rich core and a ɑ-helix-rich shell. We further demonstrate that this structural partition exists only in cells with the [RNQ+] prion state, while [rnq-] cells only carry smaller β-rich non-toxic aggregates. Collectively, our methodology has the potential to unveil detailed structural information on protein assemblies in live cells, enabling high-throughput structural screenings of macromolecular assemblies.
Collapse
Affiliation(s)
- Zhongyue Guo
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Photonics Center, Boston University, Boston, MA 02215, USA
| | - Giulio Chiesa
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Jiaze Yin
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| | - Adam Sanford
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Stefan Meier
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Ahmad S Khalil
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Ji-Xin Cheng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
3
|
Romussi S, Giunti S, Andersen N, De Rosa MJ. C. elegans: a prominent platform for modeling and drug screening in neurological disorders. Expert Opin Drug Discov 2024; 19:565-585. [PMID: 38509691 DOI: 10.1080/17460441.2024.2329103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Human neurodevelopmental and neurodegenerative diseases (NDevDs and NDegDs, respectively) encompass a broad spectrum of disorders affecting the nervous system with an increasing incidence. In this context, the nematode C. elegans, has emerged as a benchmark model for biological research, especially in the field of neuroscience. AREAS COVERED The authors highlight the numerous advantages of this tiny worm as a model for exploring nervous system pathologies and as a platform for drug discovery. There is a particular focus given to describing the existing models of C. elegans for the study of NDevDs and NDegDs. Specifically, the authors underscore their strong applicability in preclinical drug development. Furthermore, they place particular emphasis on detailing the common techniques employed to explore the nervous system in both healthy and diseased states. EXPERT OPINION Drug discovery constitutes a long and expensive process. The incorporation of invertebrate models, such as C. elegans, stands as an exemplary strategy for mitigating costs and expediting timelines. The utilization of C. elegans as a platform to replicate nervous system pathologies and conduct high-throughput automated assays in the initial phases of drug discovery is pivotal for rendering therapeutic options more attainable and cost-effective.
Collapse
Affiliation(s)
- Stefano Romussi
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
| | - Sebastián Giunti
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Natalia Andersen
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María José De Rosa
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| |
Collapse
|
4
|
Ullah A, Lee GJ, Kwon HT, Lim SI. Covalent immobilization of human serum albumin on cellulose acetate membrane for scavenging amyloid beta - A stepping extracorporeal strategy for ameliorating Alzheimer's disease. Colloids Surf B Biointerfaces 2024; 234:113753. [PMID: 38241888 DOI: 10.1016/j.colsurfb.2024.113753] [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: 10/29/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by interrupted neurocognitive functions and impaired mental development presumably caused by the accumulation of amyloid beta (Aβ) in the form of plaques. Targeting Aβ has been considered a promising approach for treating AD. In the current study, human serum albumin (HSA), a natural Aβ binder, is covalently immobilized onto the surface of a cellulose acetate (CA) membrane to devise an extracorporeal Aβ sequester. The immobilization of HSA at 3.06 ± 0.22 μg/mm2 of the CA membrane was found to be active functionally, as evidenced by the esterase-like activity converting p-nitrophenyl acetate into p-nitrophenol. The green fluorescent protein-Aβ (GFP-Aβ) fusion protein, recombinantly produced as a model ligand, exhibited characteristics of native Aβ. These features include the propensity to form aggregates or fibrils and an affinity for HSA with a dissociation constant (KD) of 0.91 μM. The HSA on the CA membrane showed concentration-dependent sequestration of GFP-Aβ in the 1-10-μM range. Moreover, it had a greater binding capacity than HSA immobilized on a commercial amine-binding plate. Results suggest that the covalent immobilization of HSA on the CA surface can be used as a potential platform for sequestering Aβ to alleviate AD.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, Khyber Pakhtunkhwa, Pakistan
| | - Gyu-Jin Lee
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Hyuk Taek Kwon
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea.
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
5
|
Schmidt SC, Massenberg A, Homsi Y, Sons D, Lang T. Microscopic clusters feature the composition of biochemical tetraspanin-assemblies and constitute building-blocks of tetraspanin enriched domains. Sci Rep 2024; 14:2093. [PMID: 38267610 PMCID: PMC10808221 DOI: 10.1038/s41598-024-52615-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/21/2024] [Indexed: 01/26/2024] Open
Abstract
Biochemical approaches revealed that tetraspanins are multi-regulatory proteins forming a web, where they act in tetraspanin-enriched-microdomains (TEMs). A microscopic criterion differentiating between web and TEMs is lacking. Using super-resolution microcopy, we identify co-assemblies between the tetraspanins CD9 and CD81 and CD151 and CD81. CD9 assemblies contain as well the CD9/CD81-interaction partner EWI-2. Moreover, CD9 clusters are proximal to clusters of the CD81-interaction partner CD44 and CD81-/EWI-2-interacting ezrin-radixin-moesin proteins. Assemblies scatter unorganized across the cell membrane; yet, upon EWI-2 elevation, they agglomerate into densely packed arranged-crowds in a process independent from actin dynamics. In conclusion, microscopic clusters are equivalent to biochemical tetraspanin-assemblies, defining in their entirety the tetraspanin web. Cluster-agglomeration enriches tetraspanins, which makes agglomerations to a microscopic complement of TEMs. The microscopic classification of tetraspanin assemblies advances our understanding of this enigmatic protein family, whose members play roles in a plethora of cellular functions, diseases, and pathogen infections.
Collapse
Affiliation(s)
- Sara C Schmidt
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Annika Massenberg
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Yahya Homsi
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Dominik Sons
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Thorsten Lang
- Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany.
| |
Collapse
|
6
|
Pasupuleti R, Riedl S, Saltor Núñez L, Karava M, Kumar V, Kourist R, Turnbull WB, Zweytick D, Wiltschi B. Lectin-anticancer peptide fusion demonstrates a significant cancer-cell-selective cytotoxic effect and inspires the production of "clickable" anticancer peptide in Escherichia coli. Protein Sci 2023; 32:e4830. [PMID: 37916438 PMCID: PMC10682692 DOI: 10.1002/pro.4830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/03/2023]
Abstract
Targeted killing of tumor cells while protecting healthy cells is the pressing priority in cancer treatment. Lectins that target a specific glycan marker abundant in cancer cells can be valuable new tools for selective cancer cell killing. The lectin Shiga-like toxin 1 B subunit (Stx1B) is an example that specifically binds globotriaosylceramide (CD77 or Gb3), which is overexpressed in certain cancers. In this study, a human lactoferricin-derived synthetic retro di-peptide R-DIM-P-LF11-215 with antitumor efficacy was fused to the lectin Stx1B to selectively target and kill Gb3+ cancer cells. We produced lectin-peptide fusion proteins in Escherichia coli, isolated them by Gb3-affinity chromatography, and assessed their ability to selectively kill Gb3+ cancer cells in a Calcein AM assay. Furthermore, to expand the applications of R-DIM-P-LF11-215 in developing therapeutic bioconjugates, we labeled R-DIM-P-LF11-215 with the unique reactive non-canonical amino acid Nε -((2-azidoethoxy)carbonyl)-L-lysine (AzK) at a selected position by amber stop codon suppression. The R-DIM-P-LF11-215 20AzK and the unlabeled R-DIM-P-LF11-215 parent peptide were produced as GST-fusion proteins for soluble expression in E. coli for the first time. We purified both variants by size-exclusion chromatography and analyzed their peptide masses. Finally, a cyanin 3 fluorophore was covalently conjugated to R-DIM-P-LF11-215 20AzK by strain-promoted alkyne-azide cycloaddition. Our results showed that the recombinant lectin-peptide fusion R-DIM-P-LF11-215-Stx1B killed >99% Gb3+ HeLa cells while Gb3-negative cells were unaffected. The peptides R-DIM-P-LF11-215 and R-DIM-P-LF11-215 20AzK were produced recombinantly in E. coli in satisfactory amounts and were tested functional by cytotoxicity and cell-binding assays, respectively.
Collapse
Affiliation(s)
- Rajeev Pasupuleti
- acib ‐ Austrian Centre of Industrial BiotechnologyGrazAustria
- Institute of Molecular BiotechnologyGraz University of TechnologyGrazAustria
| | - Sabrina Riedl
- Institute of Molecular Biosciences, Biophysics DivisionUniversity of GrazGrazAustria
- BioHealthUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| | - Laia Saltor Núñez
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsUK
| | - Marianna Karava
- Institute of Molecular BiotechnologyGraz University of TechnologyGrazAustria
| | - Vajinder Kumar
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsUK
| | - Robert Kourist
- Institute of Molecular BiotechnologyGraz University of TechnologyGrazAustria
| | - W. Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsUK
| | - Dagmar Zweytick
- Institute of Molecular Biosciences, Biophysics DivisionUniversity of GrazGrazAustria
- BioHealthUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| | - Birgit Wiltschi
- acib ‐ Austrian Centre of Industrial BiotechnologyGrazAustria
- Institute of Molecular BiotechnologyGraz University of TechnologyGrazAustria
- BioTechMed‐GrazGrazAustria
- Institute of Bioprocess Science and Engineering, Department of BiotechnologyUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
7
|
Porosk L, Härk HH, Bicev RN, Gaidutšik I, Nebogatova J, Armolik EJ, Arukuusk P, da Silva ER, Langel Ü. Aggregation Limiting Cell-Penetrating Peptides Derived from Protein Signal Sequences. Int J Mol Sci 2023; 24:ijms24054277. [PMID: 36901707 PMCID: PMC10002422 DOI: 10.3390/ijms24054277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease (ND) and the leading cause of dementia. It is characterized by non-linear, genetic-driven pathophysiological dynamics with high heterogeneity in the biological alterations and the causes of the disease. One of the hallmarks of the AD is the progression of plaques of aggregated amyloid-β (Aβ) or neurofibrillary tangles of Tau. Currently there is no efficient treatment for the AD. Nevertheless, several breakthroughs in revealing the mechanisms behind progression of the AD have led to the discovery of possible therapeutic targets. Some of these include the reduction in inflammation in the brain, and, although highly debated, limiting of the aggregation of the Aβ. In this work we show that similarly to the Neural cell adhesion molecule 1 (NCAM1) signal sequence, other Aβ interacting protein sequences, especially derived from Transthyretin, can be used successfully to reduce or target the amyloid aggregation/aggregates in vitro. The modified signal peptides with cell-penetrating properties reduce the Aβ aggregation and are predicted to have anti-inflammatory properties. Furthermore, we show that by expressing the Aβ-EGFP fusion protein, we can efficiently assess the potential for reduction in aggregation, and the CPP properties of peptides in mammalian cells.
Collapse
Affiliation(s)
- Ly Porosk
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
- Correspondence:
| | - Heleri Heike Härk
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Renata Naporano Bicev
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Ilja Gaidutšik
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | | | - Eger-Jasper Armolik
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Piret Arukuusk
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | | | - Ülo Langel
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
- Department Biochemistry and Biophysics, Stockholm University, S.Arrheniusv. 16B, Room C472, 106 91 Stockholm, Sweden
| |
Collapse
|
8
|
Wen Y, Zhang L, Li N, Tong A, Zhao C. Nutritional assessment models for Alzheimer's disease: Advances and perspectives. FOOD FRONTIERS 2023. [DOI: 10.1002/fft2.216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Affiliation(s)
- Yuxi Wen
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry Faculty of Sciences Ourense Spain
| | - Lizhu Zhang
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Na Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Aijun Tong
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
9
|
Condello C, Merz GE, Aoyagi A, DeGrado WF, Prusiner SB. Aβ and Tau Prions Causing Alzheimer's Disease. Methods Mol Biol 2023; 2561:293-337. [PMID: 36399277 DOI: 10.1007/978-1-0716-2655-9_16] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Studies show that patients with Alzheimer's disease (AD) have both Aβ and tau prions, and thus, AD is a double-prion disease. AD patients with the greatest longevity exhibited low levels of both Aβ and tau prions; tau prions were nearly absent in the brains of almost half of the patients who lived beyond 80 years of age. Using cellular bioassays for prions in postmortem samples, we found that both Aβ and tau proteins misfold into prions leading to AD, which is either a sporadic or familial dementing disorder. Although AD is transmissible experimentally, there is no evidence that AD is either communicable or contagious. Since the progression of AD correlates poorly with insoluble Aβ in the central nervous system (CNS), it was difficult to distinguish between inert amyloids and Aβ prions. To measure the progression of AD, we devised rapid bioassays to measure the abundance of isoform-specific Aβ prions in the brains of transgenic (Tg) mice and in postmortem human CNS samples from AD victims and people who died of other neurodegenerative diseases (NDs). We found significant correlations between the longevity of individuals with AD, sex, and genetic background, despite the fact that all postmortem brain tissue had essentially the same confirmed neuropathology.Although brains from all AD patients had measurable levels of Aβ prions at death, the oldest individuals had lower Aβ prion levels than the younger ones. Additionally, the long-lived individuals had low tau prion levels that correlated with the extent of phosphorylated tau (p-tau). Unexpectedly, a longevity-dependent decrease in tau prions was found in spite of increasing amounts of total insoluble tau. When corrected for the abundance of insoluble tau, the tau prion levels decreased exponentially with respect to the age at death with a half-time of approximately one decade, and this correlated with the abundance of phosphorylated tau.Even though our findings with tau prions were not unexpected, they were counterintuitive; thus, tau phosphorylation and tau prion activity decreased exponentially with longevity in patients with AD ranging from ages 37 to 99 years. Our findings demonstrated an inverse correlation between longevity in AD patients and the abundance of neurotoxic tau prions. Moreover, our discovery may have profound implications for the selection of phenotypically distinct patient populations and the development of diagnostics and effective therapeutics for AD.
Collapse
Affiliation(s)
- Carlo Condello
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| | - Gregory E Merz
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Atsushi Aoyagi
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - William F DeGrado
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
10
|
Navarro-Hortal MD, Romero-Márquez JM, Osta S, Jiménez-Trigo V, Muñoz-Ollero P, Varela-López A. Natural Bioactive Products and Alzheimer’s Disease Pathology: Lessons from Caenorhabditis elegans Transgenic Models. Diseases 2022; 10:diseases10020028. [PMID: 35645249 PMCID: PMC9149938 DOI: 10.3390/diseases10020028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-dependent, progressive disorder affecting millions of people. Currently, the therapeutics for AD only treat the symptoms. Although they have been used to discover new products of interest for this disease, mammalian models used to investigate the molecular determinants of this disease are often prohibitively expensive, time-consuming and very complex. On the other hand, cell cultures lack the organism complexity involved in AD. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for the investigation of the pathophysiology of human AD. Numerous models of both Tau- and Aβ-induced toxicity, the two prime components observed to correlate with AD pathology and the ease of performing RNA interference for any gene in the C. elegans genome, allow for the identification of multiple therapeutic targets. The effects of many natural products in main AD hallmarks using these models suggest promising health-promoting effects. However, the way in which they exert such effects is not entirely clear. One of the reasons is that various possible therapeutic targets have not been evaluated in many studies. The present review aims to explore shared therapeutical targets and the potential of each of them for AD treatment or prevention.
Collapse
|
11
|
The transmembrane domain of the amyloid precursor protein is required for anti-amyloidogenic processing by α-secretase ADAM10. J Biol Chem 2022; 298:101911. [PMID: 35398353 PMCID: PMC9127328 DOI: 10.1016/j.jbc.2022.101911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 11/24/2022] Open
Abstract
Neurotoxic amyloid β-peptides (Aβ) are thought to be a causative agent of Alzheimer's disease in humans. The production of Aβ from amyloid precursor protein (APP) could be diminished by enhancing α-processing; however, the physical interactions between APP and α-secretases are not well understood. In this study, we employed super-resolution light microscopy to examine in cell-free plasma membranes the abundance and association of APP and α-secretases ADAM10 and ADAM17. We found that both secretase molecules localize similarly closely to APP (within ≤ 50 nm). However, when cross-linking APP with antibodies directed against the GFP-tag of APP, in confocal microscopy we observed that only ADAM10 co-aggregated with APP. Furthermore, we mapped the involved protein domain by using APP variants with an exchanged transmembrane segment or lacking cytoplasmic/extracellular domains. We identified that APP's transmembrane domain is required for association with α-secretases and, as analysed by Western Blot, for α-processing. We propose that the APP transmembrane domain interacts either directly or indirectly with ADAM10, but not with ADAM17, explaining the dominant role of ADAM10 in α-processing of APP. Further understanding of this interaction may facilitate the development of a therapeutic strategy based on promoting APP cleavage by α-secretases.
Collapse
|
12
|
Utomo RY, Sugie A, Okada S, Miura K, Nakamura H. Detoxification of amyloid β fibrils by curcumin derivatives and their verification in a Drosophila Alzheimer's model. Chem Commun (Camb) 2022; 58:2576-2579. [PMID: 35107458 DOI: 10.1039/d1cc07000b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Curcumin derivatives B and N were developed as disaggregation agents of amyloid β (Aβ) fibrils. The detoxification provided by each compound at a concentration of 1 μM was observed in neuroblastoma cells. Furthermore, both compounds significantly rescued locomotion dysfunction in an Aβ-expressing Drosophila model of Alzheimer's disease.
Collapse
Affiliation(s)
- Rohmad Yudi Utomo
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan.
| | - Atsushi Sugie
- Brain Research Institute, Niigata University, 757, Ichiban-cho, Asahimachidori, Chuo-ku, Niigata 951-8585, Japan.
| | - Satoshi Okada
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan. .,Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan.,JST, PRESTO, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan
| | - Kazuki Miura
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan. .,Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan
| | - Hiroyuki Nakamura
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan. .,Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta, Midori, Yokohama, Kanagawa 226-8503, Japan
| |
Collapse
|
13
|
Modeling Alzheimer's Disease in Caenorhabditis elegans. Biomedicines 2022; 10:biomedicines10020288. [PMID: 35203497 PMCID: PMC8869312 DOI: 10.3390/biomedicines10020288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is the most frequent cause of dementia. After decades of research, we know the importance of the accumulation of protein aggregates such as β-amyloid peptide and phosphorylated tau. We also know that mutations in certain proteins generate early-onset Alzheimer’s disease (EOAD), and many other genes modulate the disease in its sporadic form. However, the precise molecular mechanisms underlying AD pathology are still unclear. Because of ethical limitations, we need to use animal models to investigate these processes. The nematode Caenorhabditis elegans has received considerable attention in the last 25 years, since the first AD models overexpressing Aβ peptide were described. We review here the main results obtained using this model to study AD. We include works studying the basic molecular mechanisms of the disease, as well as those searching for new therapeutic targets. Although this model also has important limitations, the ability of this nematode to generate knock-out or overexpression models of any gene, single or combined, and to carry out toxicity, recovery or survival studies in short timeframes with many individuals and at low cost is difficult to overcome. We can predict that its use as a model for various diseases will certainly continue to increase.
Collapse
|
14
|
Truong DJJ, Armbrust N, Geilenkeuser J, Lederer EM, Santl TH, Beyer M, Ittermann S, Steinmaßl E, Dyka M, Raffl G, Phlairaharn T, Greisle T, Živanić M, Grosch M, Drukker M, Westmeyer GG. Intron-encoded cistronic transcripts for minimally invasive monitoring of coding and non-coding RNAs. Nat Cell Biol 2022; 24:1666-1676. [PMID: 36344775 PMCID: PMC9643161 DOI: 10.1038/s41556-022-00998-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
Despite their fundamental role in assessing (patho)physiological cell states, conventional gene reporters can follow gene expression but leave scars on the proteins or substantially alter the mature messenger RNA. Multi-time-point measurements of non-coding RNAs are currently impossible without modifying their nucleotide sequence, which can alter their native function, half-life and localization. Thus, we developed the intron-encoded scarless programmable extranuclear cistronic transcript (INSPECT) as a minimally invasive transcriptional reporter embedded within an intron of a gene of interest. Post-transcriptional excision of INSPECT results in the mature endogenous RNA without sequence alterations and an additional engineered transcript that leaves the nucleus by hijacking the nuclear export machinery for subsequent translation into a reporter or effector protein. We showcase its use in monitoring interleukin-2 (IL2) after T cell activation and tracking the transcriptional dynamics of the long non-coding RNA (lncRNA) NEAT1 during CRISPR interference-mediated perturbation. INSPECT is a method for monitoring gene transcription without altering the mature lncRNA or messenger RNA of the target of interest.
Collapse
Affiliation(s)
- Dong-Jiunn Jeffery Truong
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Niklas Armbrust
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Julian Geilenkeuser
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Eva-Maria Lederer
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Tobias Heinrich Santl
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Maren Beyer
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Sebastian Ittermann
- grid.4567.00000 0004 0483 2525Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Emily Steinmaßl
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Mariya Dyka
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Gerald Raffl
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Teeradon Phlairaharn
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Tobias Greisle
- grid.4567.00000 0004 0483 2525Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Milica Živanić
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Markus Grosch
- grid.4567.00000 0004 0483 2525Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Micha Drukker
- grid.4567.00000 0004 0483 2525Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gil Gregor Westmeyer
- grid.4567.00000 0004 0483 2525Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany ,grid.6936.a0000000123222966Department of Chemistry and TUM School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
15
|
Shillcock JC, Hastings J, Riguet N, Lashuel HA. Non-monotonic fibril surface occlusion by GFP tags from coarse-grained molecular simulations. Comput Struct Biotechnol J 2021; 20:309-321. [PMID: 35070162 PMCID: PMC8753129 DOI: 10.1016/j.csbj.2021.12.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 11/23/2022] Open
Abstract
The pathological growth of amyloid fibrils in neurons underlies the progression of neurodegenerative diseases including Alzheimer's and Parkinson's disease. Fibrils form when soluble monomers oligomerise in the cytoplasm. Their subsequent growth occurs via nucleated polymerization mechanisms involving the free ends of the fibrils augmented by secondary nucleation of new oligomers at their surface. Amyloid fibrils possess a complex interactome with diffusing cytoplasmic proteins that regulates many aspects of their growth, seeding capacity, biochemical activity and transition to pathological inclusions in diseased brains. Changes to their surface are also expected to modify their interactome, pathogenicity and spreading in the brain. Many assays visualise fibril formation, growth and inclusion formation by decorating monomeric proteins with fluorescent tags such as GFP. Recent studies from our group suggest that tags with sizes comparable to the fibril radius may modify the fibril surface accessibility and thus their PTM pattern, interactome and ability to form inclusions. Using coarse-grained molecular simulations of a single alpha synuclein fibril tagged with GFP we find that thermal fluctuations of the tags create a non-monotonic, size-dependent sieve around the fibril that perturbs its interactome with diffusing species. Our results indicate that experiments using tagged and untagged monomers to study the growth and interactome of fibrils should be compared with caution, and the confounding effects of the tags are more complex than a reduction in surface accessibility. The prevalence of fluorescent tags in amyloid fibril growth experiments suggests this has implications beyond the specific alpha synuclein fibrils we model here.
Collapse
Affiliation(s)
- Julian C. Shillcock
- Blue Brain Project, Ecole polytechnique fédérale de Lausanne, CH-1202 Geneva, Switzerland
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole polytechnique fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Janna Hastings
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole polytechnique fédérale de Lausanne, CH-1015 Lausanne, Switzerland
- Bioinformatics Competence Center, Ecole polytechnique fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Nathan Riguet
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole polytechnique fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Ecole polytechnique fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
16
|
Mertins J, Finke J, Sies R, Rink KM, Hasenauer J, Lang T. The mesoscale organization of syntaxin 1A and SNAP25 is determined by SNARE-SNARE interactions. eLife 2021; 10:69236. [PMID: 34779769 PMCID: PMC8629428 DOI: 10.7554/elife.69236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 11/14/2021] [Indexed: 01/01/2023] Open
Abstract
SNARE proteins have been described as the effectors of fusion events in the secretory pathway more than two decades ago. The strong interactions between SNARE domains are clearly important in membrane fusion, but it is unclear whether they are involved in any other cellular processes. Here, we analyzed two classical SNARE proteins, syntaxin 1A and SNAP25. Although they are supposed to be engaged in tight complexes, we surprisingly find them largely segregated in the plasma membrane. Syntaxin 1A only occupies a small fraction of the plasma membrane area. Yet, we find it is able to redistribute the far more abundant SNAP25 on the mesoscale by gathering crowds of SNAP25 molecules onto syntaxin clusters in a SNARE-domain-dependent manner. Our data suggest that SNARE domain interactions are not only involved in driving membrane fusion on the nanoscale, but also play an important role in controlling the general organization of proteins on the mesoscale. Further, we propose these mechanisms preserve active syntaxin 1A–SNAP25 complexes at the plasma membrane.
Collapse
Affiliation(s)
- Jasmin Mertins
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jérôme Finke
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Ricarda Sies
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Kerstin M Rink
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Jan Hasenauer
- Computational Life Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Interdisciplinary Research Unit Mathematics and Life Sciences, University of Bonn, Bonn, Germany.,Institute of Computational Biology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thorsten Lang
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
17
|
Good SC, Dewison KM, Radford SE, van Oosten-Hawle P. Global Proteotoxicity Caused by Human β 2 Microglobulin Variants Impairs the Unfolded Protein Response in C. elegans. Int J Mol Sci 2021; 22:10752. [PMID: 34639093 PMCID: PMC8509642 DOI: 10.3390/ijms221910752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022] Open
Abstract
Aggregation of β2 microglobulin (β2m) into amyloid fibrils is associated with systemic amyloidosis, caused by the deposition of amyloid fibrils containing the wild-type protein and its truncated variant, ΔN6 β2m, in haemo-dialysed patients. A second form of familial systemic amyloidosis caused by the β2m variant, D76N, results in amyloid deposits in the viscera, without renal dysfunction. Although the folding and misfolding mechanisms of β2 microglobulin have been widely studied in vitro and in vivo, we lack a comparable understanding of the molecular mechanisms underlying toxicity in a cellular and organismal environment. Here, we established transgenic C. elegans lines expressing wild-type (WT) human β2m, or the two highly amyloidogenic naturally occurring variants, D76N β2m and ΔN6 β2m, in the C. elegans bodywall muscle. Nematodes expressing the D76N β2m and ΔN6 β2m variants exhibit increased age-dependent and cell nonautonomous proteotoxicity associated with reduced motility, delayed development and shortened lifespan. Both β2m variants cause widespread endogenous protein aggregation contributing to the increased toxicity in aged animals. We show that expression of β2m reduces the capacity of C. elegans to cope with heat and endoplasmic reticulum (ER) stress, correlating with a deficiency to upregulate BiP/hsp-4 transcripts in response to ER stress in young adult animals. Interestingly, protein secretion in all β2m variants is reduced, despite the presence of the natural signal sequence, suggesting a possible link between organismal β2m toxicity and a disrupted ER secretory metabolism.
Collapse
Affiliation(s)
| | | | | | - Patricija van Oosten-Hawle
- Faculty of Biological Sciences, School of Molecular and Cell Biology & Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK; (S.C.G.); (K.M.D.); (S.E.R.)
| |
Collapse
|
18
|
Ochiishi T, Kaku M, Kajsongkram T, Thisayakorn K. Mulberry fruit extract alleviates the intracellular amyloid-β oligomer-induced cognitive disturbance and oxidative stress in Alzheimer's disease model mice. Genes Cells 2021; 26:861-873. [PMID: 34387016 DOI: 10.1111/gtc.12889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/30/2022]
Abstract
Intracellular amyloid-β (Aβ) oligomers are key therapeutic targets because they are strongly cytotoxic and play crucial roles in the cognitive function in Alzheimer's disease (AD). Anthocyanins, polyphenolic flavonoids with antioxidant and neuroprotective properties, are potential therapeutic candidates for AD. Here, we investigated the effects of anthocyanin-enriched extracts from fruits of mulberry (Morus alba Linn.) in Thailand against the neurotoxicity of Aβ oligomers. Using the monitoring system for Aβ aggregation, we showed that the extract induced the dissociation of Aβ in cultured HEK293T cells. To investigate the effects on cognitive function, we orally administered the extract to Aβ-GFP transgenic mice (Aβ-GFP Tg), a mouse model that expresses Aβ oligomers inside neurons, and performed the novel object recognition test and passive avoidance test. Aβ-GFP Tg usually showed deficits in novel object recognition memory and reference memory compared with non-Tg, but administration of the extract improved both compared with vehicle-treated Aβ-GFP Tg. Aβ-GFP Tg exhibited lower superoxide dismutase (SOD) activity than non-Tg. However, after the administration of the extract, the SOD activity was restored. These results suggest that Thai mulberry fruit extract ameliorates cytotoxicity induced by the intracellular Aβ oligomers and may be an effective therapeutic or preventive candidate for AD.
Collapse
Affiliation(s)
- Tomoyo Ochiishi
- Molecular Neurobiology Research Group, Biomedical Research Institute (BMRI), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Masami Kaku
- Faculty of Health and Science, Uekusa Gakuen University, Chiba, Japan
| | - Tanwarat Kajsongkram
- Expert Centre of Innovative Herbal Products (InnoHerb), Thailand Institute of Scientific and Technological Research (TISTR), Khlong Luang, Thailand
| | - Krittiya Thisayakorn
- Expert Centre of Innovative Herbal Products (InnoHerb), Thailand Institute of Scientific and Technological Research (TISTR), Khlong Luang, Thailand
| |
Collapse
|
19
|
Khan AN, Gadhave K, Furkan M, Kumar P, Siddiqi MK, Giri R, Khan RH. Anti-tuberculotic thionamide antibiotics show antioxidative and neuronal cytoprotective nature by inhibiting amyloid formation in human insulin and amyloid β-42. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.115396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Azizyan RA, Wang W, Anikeenko A, Radkova Z, Bakulina A, Garro A, Charlier L, Dumas C, Ventura S, Kajava AV. Amyloidogenicity as a driving force for the formation of functional oligomers. J Struct Biol 2020; 212:107604. [PMID: 32805411 DOI: 10.1016/j.jsb.2020.107604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 11/28/2022]
Abstract
Insoluble amyloid fibrils formed by self-assembly of amyloidogenic regions of proteins have a cross-β-structure. In this work, by using targeted molecular dynamics and rigid body simulation, we demonstrate that if a protein consists of an amyloidogenic region and a globular domain(s) and if the linker between them is short enough, such molecules cannot assemble into amyloid fibrils, instead, they form oligomers with a defined and limited number of β-strands in the cross-β core. We show that this blockage of the amyloid growth is due to the steric repulsion of the globular structures linked to amyloidogenic regions. Furthermore, we establish a relationship between the linker length and the number of monomers in such nanoparticles. We hypothesise that such oligomerisation can be a yet unrecognised way to form natural protein complexes involved in biological processes. Our results can also be used in protein engineering for designing soluble nanoparticles carrying different functional domains.
Collapse
Affiliation(s)
- Rafayel A Azizyan
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237, CNRS, Université Montpellier, Montpellier, France; Institut de Biologie Computationnelle, Université Montpellier, Montpellier, France
| | - Weiqiang Wang
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193-Bellaterra, Spain
| | | | | | | | - Adriana Garro
- Universidad Nacional de San Luis IMASL-CONICET, San Luis, Argentina
| | - Landry Charlier
- Institut des Biomolécules Max Mousseron, Montpellier, France
| | - Christian Dumas
- Centre de Biochimie Structurale, CNRS, UMR5048, INSERM, U1054, Université de Montpellier, Montpellier, France
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193-Bellaterra, Spain
| | - Andrey V Kajava
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237, CNRS, Université Montpellier, Montpellier, France; Institut de Biologie Computationnelle, Université Montpellier, Montpellier, France.
| |
Collapse
|
21
|
Gallrein C, Iburg M, Michelberger T, Koçak A, Puchkov D, Liu F, Ayala Mariscal SM, Nayak T, Kaminski Schierle GS, Kirstein J. Novel amyloid-beta pathology C. elegans model reveals distinct neurons as seeds of pathogenicity. Prog Neurobiol 2020; 198:101907. [PMID: 32926945 DOI: 10.1016/j.pneurobio.2020.101907] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/28/2020] [Accepted: 09/01/2020] [Indexed: 11/30/2022]
Abstract
Protein misfolding and aggregation are hallmarks of neurodegenerative diseases such as Alzheimer's disease (AD). In AD, the accumulation and aggregation of tau and the amyloid-beta peptide Aβ1-42 precedes the onset of AD symptoms. Modelling the aggregation of Aβ is technically very challenging in vivo due to its size of only 42 aa. Here, we employed sub-stoichiometric labelling of Aβ1-42 in C. elegans to enable tracking of the peptide in vivo, combined with the "native" aggregation of unlabeled Aβ1-42. Expression of Aβ1-42 leads to severe physiological defects, neuronal dysfunction and neurodegeneration. Moreover, we can demonstrate spreading of neuronal Aβ to other tissues. Fluorescence lifetime imaging microscopy enabled a quantification of the formation of amyloid fibrils with ageing and revealed a heterogenic yet specific pattern of aggregation. Notably, we found that Aβ aggregation starts in a subset of neurons of the anterior head ganglion, the six IL2 neurons. We further demonstrate that cell-specific, RNAi-mediated depletion of Aβ in these IL2 neurons systemically delays Aβ aggregation and pathology.
Collapse
Affiliation(s)
- Christian Gallrein
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Manuel Iburg
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Tim Michelberger
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Alen Koçak
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Dmytro Puchkov
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Fan Liu
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Sara Maria Ayala Mariscal
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Tanmoyita Nayak
- University of Bremen, Faculty 2, Cell Biology, Leobener Strasse, 28359, Bremen, Germany
| | - Gabriele S Kaminski Schierle
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Janine Kirstein
- Leibniz Research Institute for Molecular Pharmacology im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, Berlin, 13125, Germany; University of Bremen, Faculty 2, Cell Biology, Leobener Strasse, 28359, Bremen, Germany.
| |
Collapse
|
22
|
Kaniyappan S, Tepper K, Biernat J, Chandupatla RR, Hübschmann S, Irsen S, Bicher S, Klatt C, Mandelkow EM, Mandelkow E. FRET-based Tau seeding assay does not represent prion-like templated assembly of Tau filaments. Mol Neurodegener 2020; 15:39. [PMID: 32677995 PMCID: PMC7364478 DOI: 10.1186/s13024-020-00389-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/08/2020] [Indexed: 01/15/2023] Open
Abstract
Tau aggregation into amyloid fibers based on the cross-beta structure is a hallmark of several Tauopathies, including Alzheimer Disease (AD). Trans-cellular propagation of Tau with pathological conformation has been suggested as a key disease mechanism. This is thought to cause the spreading of Tau pathology in AD by templated conversion of naive Tau in recipient cells into a pathological state, followed by assembly of pathological Tau fibers, similar to the mechanism of nucleated polymerization proposed for prion pathogenesis. In cell cultures, the process is often monitored by a FRET assay where the recipient cell expresses the Tau repeat domain (TauRD) with a pro-aggregant mutation, fused to GFP-based FRET pairs. Since the size of the reporter GFP (barrel of ~ 3 nm × 4 nm) is ~ 7 times larger than the β-strand distance (0.47 nm), this points to a potential steric clash. Hence, we investigated the influence of the GFP tag on TauFL or TauRD aggregation. Using biophysical methods (light scattering, atomic force microscopy (AFM), and scanning-transmission electron microscopy (STEM)), we found that the assembly of TauRD-GFP was severely inhibited and incompatible with that of Alzheimer filaments. These observations argue against the hypothesis that the propagation of Tau pathology in AD is caused by the prion-like templated aggregation of Tau protein, transmitted via cell-to-cell spreading of Tau. Thus, even though the observed local increase of FRET in recipient cells may be a valid hallmark of a pathological reaction, our data argue that it is caused by a process distinct from assembly of TauRD filaments.
Collapse
Affiliation(s)
- Senthilvelrajan Kaniyappan
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany. .,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany.
| | - Katharina Tepper
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Jacek Biernat
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | | | | | | | | | - Eva-Maria Mandelkow
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany.,CAESAR Research Center, Bonn, Germany
| | - Eckhard Mandelkow
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany. .,CAESAR Research Center, Bonn, Germany.
| |
Collapse
|
23
|
Aoyagi A, Condello C, Stöhr J, Yue W, Rivera BM, Lee JC, Woerman AL, Halliday G, van Duinen S, Ingelsson M, Lannfelt L, Graff C, Bird TD, Keene CD, Seeley WW, DeGrado WF, Prusiner SB. Aβ and tau prion-like activities decline with longevity in the Alzheimer's disease human brain. Sci Transl Med 2020; 11:11/490/eaat8462. [PMID: 31043574 DOI: 10.1126/scitranslmed.aat8462] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022]
Abstract
The hallmarks of Alzheimer's disease (AD) are the accumulation of Aβ plaques and neurofibrillary tangles composed of hyperphosphorylated tau. We developed sensitive cellular assays using human embryonic kidney-293T cells to quantify intracellular self-propagating conformers of Aβ in brain samples from patients with AD or other neurodegenerative diseases. Postmortem brain tissue from patients with AD had measurable amounts of pathological Aβ conformers. Individuals over 80 years of age had the lowest amounts of prion-like Aβ and phosphorylated tau. Unexpectedly, the longevity-dependent decrease in self-propagating tau conformers occurred in spite of increasing amounts of total insoluble tau. When corrected for the abundance of insoluble tau, the ability of postmortem AD brain homogenates to induce misfolded tau in the cellular assays showed an exponential decrease with longevity, with a half-life of about one decade over the age range of 37 to 99 years. Thus, our findings demonstrate an inverse correlation between longevity in patients with AD and the abundance of pathological tau conformers. Our cellular assays can be applied to patient selection for clinical studies and the development of new drugs and diagnostics for AD.
Collapse
Affiliation(s)
- Atsushi Aoyagi
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Daiichi Sankyo Co. Ltd., Tokyo 140-8710, Japan
| | - Carlo Condello
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA. .,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jan Stöhr
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Weizhou Yue
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Brianna M Rivera
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Joanne C Lee
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Amanda L Woerman
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Glenda Halliday
- NeuRA and School of Medical Sciences, University of New South Wales, and Brain and Mind Centre, University of Sydney, Sydney, NSW 2052, Australia
| | | | - Martin Ingelsson
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, 751 85 Uppsala, Sweden
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, 751 85 Uppsala, Sweden
| | - Caroline Graff
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden.,Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Thomas D Bird
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA.,Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - C Dirk Keene
- Department of Neuropathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - William W Seeley
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William F DeGrado
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA. .,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
24
|
Jin Y, Chung YW, Jung MK, Lee JH, Ko KY, Jang JK, Ham M, Kang H, Pack CG, Mihara H, Kim IY. Apolipoprotein E-mediated regulation of selenoprotein P transportation via exosomes. Cell Mol Life Sci 2020; 77:2367-2386. [PMID: 31471680 PMCID: PMC11104972 DOI: 10.1007/s00018-019-03287-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/14/2019] [Accepted: 08/23/2019] [Indexed: 10/26/2022]
Abstract
Selenoprotein P (SELENOP), secreted from the liver, functions as a selenium (Se) supplier to other tissues. In the brain, Se homeostasis is critical for physiological function. Previous studies have reported that SELENOP co-localizes with the apolipoprotein E receptor 2 (ApoER2) along the blood-brain barrier (BBB). However, the mechanism underlying SELENOP transportation from hepatocytes to neuronal cells remains unclear. Here, we found that SELENOP was secreted from hepatocytes as an exosomal component protected from plasma kallikrein-mediated cleavage. SELENOP was interacted with apolipoprotein E (ApoE) through heparin-binding sites of SELENOP, and the interaction regulated the secretion of exosomal SELENOP. Using in vitro BBB model of transwell cell culture, exosomal SELENOP was found to supply Se to brain endothelial cells and neuronal cells, which synthesized selenoproteins by a process regulated by ApoE and ApoER2. The regulatory role of ApoE in SELENOP transport was also observed in vivo using ApoE-/- mice. Exosomal SELENOP transport protected neuronal cells from amyloid β (Aβ)-induced cell death. Taken together, our results suggest a new delivery mechanism for Se to neuronal cells by exosomal SELENOP.
Collapse
Affiliation(s)
- Yunjung Jin
- Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Youn Wook Chung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Min Kyo Jung
- Asan Institute for Life Sciences, Asan Medical Center & Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
- Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, 41068, South Korea
| | - Jea Hwang Lee
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts General Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, 02114, USA
| | - Kwan Young Ko
- Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Jun Ki Jang
- Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Minju Ham
- Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunwoo Kang
- Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Chan Gi Pack
- Asan Institute for Life Sciences, Asan Medical Center & Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Hisaaki Mihara
- Department of Biotechnology, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Ick Young Kim
- Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
25
|
Watanabe C, Yanagisawa M. Unique phase behavior in cell size space: synergistic effect of molecular crowding and confinement. Biophys Rev 2020; 12:385-386. [PMID: 32277345 DOI: 10.1007/s12551-020-00656-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/18/2020] [Indexed: 01/19/2023] Open
Affiliation(s)
- Chiho Watanabe
- Komaba Institute for Science, The University of Tokyo, Bunkyo, Japan
| | - Miho Yanagisawa
- Komaba Institute for Science, The University of Tokyo, Bunkyo, Japan.
- Department of Basic Science, The University of Tokyo, Bunkyo, Japan.
| |
Collapse
|
26
|
Watanabe C, Kobori Y, Yamamoto J, Kinjo M, Yanagisawa M. Quantitative Analysis of Membrane Surface and Small Confinement Effects on Molecular Diffusion. J Phys Chem B 2020; 124:1090-1098. [PMID: 31939302 DOI: 10.1021/acs.jpcb.9b10558] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Molecular behaviors in small liquid droplets (picoliter scale), such as phase transitions and chemical reactions, are essential for the industrial application of small droplets and their use as artificial cells. However, the droplets often differ from those in bulk solutions (milliliter scale). Since the droplet size is much larger than the molecular size, the so-called size effect that draws these differences has attracted attention as a target to be solved. Although the small volume and the membrane surface surrounding the droplet are thought to be the origin of the size effect, there were little attempts to separate and quantify them. To solve the problem, we develop a series of systems for the evaluation. Using these systems, we have evaluated the size effect of concentrated polymer solutions on molecular diffusion by dividing it into small volume and membrane surface contributions. Our results demonstrate that the size effect on the molecular diffusion originates from the long-range interaction with the surface enhanced with decreasing volume. The quantitative size effect revealed by the systems provides novel insights in the biophysical understanding of molecular behaviors in cells and to the regulation and design of micrometer-sized materials.
Collapse
Affiliation(s)
- Chiho Watanabe
- Komaba Institute for Science , The University of Tokyo , Komaba 3-8-1 , Meguro , Tokyo 153-8902 , Japan
| | - Yuta Kobori
- Komaba Institute for Science , The University of Tokyo , Komaba 3-8-1 , Meguro , Tokyo 153-8902 , Japan.,Department of Applied Physics , Tokyo University of Agriculture and Technology , Naka-cho 2-24-16 , Koganei , Tokyo 184-8588 , Japan
| | - Johtaro Yamamoto
- Biomedical Research Institute , National Institute of Advanced Industrial Science and Technology (AIST) , Central 6, Higashi 1-1-1 , Tsukuba , Ibaraki 305-8568 , Japan
| | - Masataka Kinjo
- Faculty of Advanced Life Science , Hokkaido University , Kita-21 Nishi-11 Kita-ku , Sapporo , Hokkaido 001-0021 , Japan
| | - Miho Yanagisawa
- Komaba Institute for Science , The University of Tokyo , Komaba 3-8-1 , Meguro , Tokyo 153-8902 , Japan.,Department of Basic Science , The University of Tokyo , Komaba 3-8-1 , Meguro , Tokyo 153-8902 , Japan
| |
Collapse
|
27
|
New Alzheimer's disease model mouse specialized for analyzing the function and toxicity of intraneuronal Amyloid β oligomers. Sci Rep 2019; 9:17368. [PMID: 31757975 PMCID: PMC6874556 DOI: 10.1038/s41598-019-53415-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/29/2019] [Indexed: 11/11/2022] Open
Abstract
Oligomers of intracellular amyloid β protein (Aβ) are strongly cytotoxic and play crucial roles in synaptic transmission and cognitive function in Alzheimer’s disease (AD). However, there is currently no AD model mouse in which to specifically analyze the function of Aβ oligomers only. We have now developed a novel AD model mouse, an Aβ-GFP transgenic mouse (Aβ-GFP Tg), that expresses the GFP-fused human Aβ1-42 protein, which forms only Aβ oligomers within neurons throughout their life. The fusion proteins are expressed mainly in the hippocampal CA1-CA2 region and cerebral cortex, and are not secreted extracellularly. The Aβ-GFP Tg mice exhibit increased tau phosphorylation, altered spine morphology, decreased expressions of the GluN2B receptor and neuroligin in synaptic regions, attenuated hippocampal long-term potentiation, and impaired object recognition memory compared with non-Tg littermates. Interestingly, these dysfunctions have already appeared in 2–3-months-old animals. The Aβ-GFP fusion protein is bioactive and highly toxic, and induces the similar synaptic dysfunctions as the naturally generated Aβ oligomer derived from postmortem AD patient brains and synthetic Aβ oligomers. Thus, Aβ-GFP Tg mouse is a new tool specialized to analyze the function of Aβ oligomers in vivo and to find subtle changes in synapses in early symptoms of AD.
Collapse
|
28
|
Developing Trojan horses to induce, diagnose and suppress Alzheimer’s pathology. Pharmacol Res 2019; 149:104471. [DOI: 10.1016/j.phrs.2019.104471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 01/05/2023]
|
29
|
Establishment of Constraints on Amyloid Formation Imposed by Steric Exclusion of Globular Domains. J Mol Biol 2018; 430:3835-3846. [DOI: 10.1016/j.jmb.2018.05.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/15/2018] [Accepted: 05/27/2018] [Indexed: 11/18/2022]
|
30
|
Shen P, Yue Y, Zheng J, Park Y. Caenorhabditis elegans: A Convenient In Vivo Model for Assessing the Impact of Food Bioactive Compounds on Obesity, Aging, and Alzheimer's Disease. Annu Rev Food Sci Technol 2018; 9:1-22. [DOI: 10.1146/annurev-food-030117-012709] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Peiyi Shen
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
31
|
State-of-the-Art Fluorescence Fluctuation-Based Spectroscopic Techniques for the Study of Protein Aggregation. Int J Mol Sci 2018; 19:ijms19040964. [PMID: 29570669 PMCID: PMC5979297 DOI: 10.3390/ijms19040964] [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: 02/20/2018] [Revised: 03/17/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease, are devastating proteinopathies with misfolded protein aggregates accumulating in neuronal cells. Inclusion bodies of protein aggregates are frequently observed in the neuronal cells of patients. Investigation of the underlying causes of neurodegeneration requires the establishment and selection of appropriate methodologies for detailed investigation of the state and conformation of protein aggregates. In the current review, we present an overview of the principles and application of several methodologies used for the elucidation of protein aggregation, specifically ones based on determination of fluctuations of fluorescence. The discussed methods include fluorescence correlation spectroscopy (FCS), imaging FCS, image correlation spectroscopy (ICS), photobleaching ICS (pbICS), number and brightness (N&B) analysis, super-resolution optical fluctuation imaging (SOFI), and transient state (TRAST) monitoring spectroscopy. Some of these methodologies are classical protein aggregation analyses, while others are not yet widely used. Collectively, the methods presented here should help the future development of research not only into protein aggregation but also neurodegenerative diseases.
Collapse
|
32
|
Shi JM, Pei J, Liu EQ, Zhang L. Bis(sulfosuccinimidyl) suberate (BS3) crosslinking analysis of the behavior of amyloid-β peptide in solution and in phospholipid membranes. PLoS One 2017; 12:e0173871. [PMID: 28323849 PMCID: PMC5360245 DOI: 10.1371/journal.pone.0173871] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 02/28/2017] [Indexed: 12/31/2022] Open
Abstract
The structure and state of amyloid-β peptide (Aβ) oligomers often need to be checked by reliable experimental methods. Electrophoresis is a commonly applied measurement method. However, due to the presence of detergents, oligomers are easily broken during electrophoresis, which makes it very hard to accurately assess Aβ aggregate states. In the current study, bis(sulfosuccinimidyl) suberate (BS3) was used to cross-link Aβ1-42 oligomers prior to electrophoresis. When compared to a previously reported Aβ cross-linking agent, glutaraldehyde, it was quite apparent that BS3 is more suitable for detecting intra-membrane Aβ oligomers and extra-membrane Aβ oligomers states. As such, our findings provide an efficient method for analyzing Aβ proteins or other proteins that are easily aggregated in solution and in phospholipid membranes.
Collapse
Affiliation(s)
- Jing-Ming Shi
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an P. R. China
- School of Medicine, Xizang Minzu University, Xian’ yang P. R. China
| | - Jie Pei
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou P. R. China
| | - En-Qi Liu
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an P. R. China
| | - Lin Zhang
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an P. R. China
| |
Collapse
|