1
|
Baysah CZ, Dohoney RA, Palanikumar L, Stillman NH, Penney AL, Sola AD, Paredes DA, Magzoub M, Kumar S. A Brain-Penetrating Foldamer Rescues Aβ Aggregation-Associated Alzheimer's Disease Phenotypes in In Vivo Models. ACS Chem Neurosci 2025; 16:1309-1322. [PMID: 40070152 DOI: 10.1021/acschemneuro.4c00753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is the leading cause of dementia, affecting nearly 55 million people across the world. One of the central pathological factors associated with AD is the aggregation of Aβ42, a peptide product cleaved through pathological processes in AD. Under pathological conditions, Aβ42 aggregates into insoluble plaques in the brain and impairs the function of neurons, which contributes to the cognitive decline associated with AD. Therefore, the modulation of Aβ42 aggregation is considered a potential therapeutic intervention for AD. Using an Oligoquinoline-based foldamer library, we have identified a potent foldamer antagonist (SK-131) of Aβ42 aggregation. SK-131 inhibits the aggregation by inducing a α-helical structure in monomeric Aβ42. Here, we demonstrated that SK-131 rescues Aβ42 aggregation-associated phenotypes in AD cellular and multiple Caenorhabditis elegans AD models, including intracellular inhibition of Aβ42 aggregation, rescue of behavioral deficits, and attenuation of reactive oxygen species. It efficiently crosses the blood-brain barrier and demonstrates favorable pharmaceutical properties. It also potently inhibits Zn2+-mediated Aβ42 aggregation by potentially displacing Zn2+ from Aβ42. In summary, we have identified a potent brain-penetrating foldamer that efficiently rescues AD phenotypes in in vivo models. Unlike most of the therapeutic approaches that target Aβ aggregates, we have identified and validated a novel therapeutic pathway by inducing structural change in Aβ and rescuing AD phenotypes in in vivo models. This study will further aid in the quest to identify lead therapeutics to slow or stop the progression of AD.
Collapse
Affiliation(s)
- Charles Zuwu Baysah
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - Ryan A Dohoney
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - L Palanikumar
- Biology Program, Division of Science, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE
| | - Nicholas H Stillman
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - Alexandra L Penney
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
- Department of Biological Sciences, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
| | - Andres D Sola
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - Daniel A Paredes
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- Ritchie School of Engineering and Computer Science, University of Denver, 2155 E Wesley Ave, Denver, Colorado 80210, United States
| | - Mazin Magzoub
- Biology Program, Division of Science, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE
| | - Sunil Kumar
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
- Molecular and Cellular Biophysics Program, University of Denver, Boettcher West, Room 228, 2050 E. Iliff Ave, Denver, Colorado 80210, United States
| |
Collapse
|
2
|
Valdés A, Sánchez-Martínez JD, Gallego R, Ibáñez E, Herrero M, Cifuentes A. In vivo neuroprotective capacity of a Dunaliella salina extract - comprehensive transcriptomics and metabolomics study. NPJ Sci Food 2024; 8:4. [PMID: 38200022 PMCID: PMC10782027 DOI: 10.1038/s41538-023-00246-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
In this study, an exhaustive chemical characterization of a Dunaliella salina (DS) microalga extract obtained using supercritical fluids has been performed, and its neuroprotective capacity has been evaluated in vivo using an Alzheimer's disease (AD) transgenic model of Caenorhabditis elegans (strain CL4176). More than 350 compounds were annotated in the studied DS extract, with triacylglycerols, free fatty acids (FAs), carotenoids, apocarotenoids and glycerol being the most abundant. DS extract significantly protects C. elegans in a dose-dependent manner against Aβ-peptide paralysis toxicity, after 32 h, 53% of treated worms at 50 µg/mL were not paralyzed. This concentration was selected to further evaluate the transcriptomics and metabolomics changes after 26 h by using advanced analytical methodologies. The RNA-Seq data showed an alteration of 150 genes, mainly related to the stress and detoxification responses, and the retinol and lipid metabolism. The comprehensive metabolomics and lipidomics analyses allowed the identification of 793 intracellular metabolites, of which 69 were significantly altered compared to non-treated control animals. Among them, different unsaturated FAs, lysophosphatidylethanolamines, nucleosides, dipeptides and modified amino acids that have been previously reported as beneficial during AD progression, were assigned. These compounds could explain the neuroprotective capacity observed, thus, providing with new evidences of the protection mechanisms of this promising extract.
Collapse
Affiliation(s)
- Alberto Valdés
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC-UAM), Calle Nicolás Cabrera 9, 28049, Madrid, Spain.
| | - José David Sánchez-Martínez
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC-UAM), Calle Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Rocío Gallego
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC-UAM), Calle Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Elena Ibáñez
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC-UAM), Calle Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Miguel Herrero
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC-UAM), Calle Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC-UAM), Calle Nicolás Cabrera 9, 28049, Madrid, Spain
| |
Collapse
|
3
|
Ayuda-Durán B, Garzón-García L, González-Manzano S, Santos-Buelga C, González-Paramás AM. Insights into the Neuroprotective Potential of Epicatechin: Effects against Aβ-Induced Toxicity in Caenorhabditis elegans. Antioxidants (Basel) 2024; 13:79. [PMID: 38247503 PMCID: PMC10812808 DOI: 10.3390/antiox13010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Medical therapies to avoid the progression of Alzheimer's disease (AD) are limited to date. Certain diets have been associated with a lower incidence of neurodegenerative diseases. In particular, the regular intake of foods rich in polyphenols, such as epicatechin (EC), could help prevent or mitigate AD progression. This work aims to explore the neuroprotective effects of EC using different transgenic strains of Caenorhabditis elegans, which express human Aβ1-42 peptides and contribute to elucidating the mechanisms involved in the effects of EC in AD. The performed assays indicate that this flavan-3-ol was able to reduce the signs of β-amyloid accumulation in C. elegans, improving motility and chemotaxis and increasing survival in transgenic strain peptide producers compared to nematodes not treated with EC. The neuroprotective effects exhibited by EC in C. elegans could be explained by the modulation of inflammation and stress-associated genes, as well as autophagy, microgliosis, and heat shock signaling pathways, involving the regulation of cpr-5, epg-8, ced-7, ZC239.12, and hsp-16 genes. Overall, the results obtained in this study support the protective effects of epicatechin against Aβ-induced toxicity.
Collapse
Affiliation(s)
| | | | | | - Celestino Santos-Buelga
- Grupo de Investigación en Polifenoles (GIP-USAL), Campus Miguel de Unamuno, Universidad de Salamanca, 37007 Salamanca, Spain; (B.A.-D.); (L.G.-G.); (S.G.-M.)
| | - Ana M. González-Paramás
- Grupo de Investigación en Polifenoles (GIP-USAL), Campus Miguel de Unamuno, Universidad de Salamanca, 37007 Salamanca, Spain; (B.A.-D.); (L.G.-G.); (S.G.-M.)
| |
Collapse
|
4
|
Jiang Y, MacNeil LT. Simple model systems reveal conserved mechanisms of Alzheimer's disease and related tauopathies. Mol Neurodegener 2023; 18:82. [PMID: 37950311 PMCID: PMC10638731 DOI: 10.1186/s13024-023-00664-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023] Open
Abstract
The lack of effective therapies that slow the progression of Alzheimer's disease (AD) and related tauopathies highlights the need for a more comprehensive understanding of the fundamental cellular mechanisms underlying these diseases. Model organisms, including yeast, worms, and flies, provide simple systems with which to investigate the mechanisms of disease. The evolutionary conservation of cellular pathways regulating proteostasis and stress response in these organisms facilitates the study of genetic factors that contribute to, or protect against, neurodegeneration. Here, we review genetic modifiers of neurodegeneration and related cellular pathways identified in the budding yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans, and the fruit fly Drosophila melanogaster, focusing on models of AD and related tauopathies. We further address the potential of simple model systems to better understand the fundamental mechanisms that lead to AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuwei Jiang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
5
|
Sánchez-Martínez JD, Cifuentes A, Valdés A. Omics approaches to investigate the neuroprotective capacity of a Citrus sinensis (sweet orange) extract in a Caenorhabditis elegans Alzheimer's model. Food Res Int 2023; 172:113128. [PMID: 37689893 DOI: 10.1016/j.foodres.2023.113128] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 09/11/2023]
Abstract
Citrus sinensis by-products are a promising source of neuroprotective molecules. In this study, a pressurized liquid extract of Citrus by-products (PLE100) has been extensively characterized, and its neuroprotective capacity tested in the Caenorhabditis elegans strain CL4176, a validated in vivo model of Alzheimer's disease (AD). More than 450 compounds have been annotated in the extract, being triacylglycerols (TGs), stigmastanes, fatty acids (FAs) and carbohydrates the most abundant. The results demonstrate that worms PLE100-treated are significantly protected in a dose-dependent manner against the Aβ-peptide paralysis toxicity. The RNA-Seq data showed an alteration of 294 genes mainly related to the stress response defense along with genes involved in the lipid transport and metabolism. Moreover, the comprehensive metabolomics study allowed the identification of 818 intracellular metabolites, of which 54 were significantly altered (mainly lipids). The integration of these and previous results provides with new evidences of the protection mechanisms of this promising extract.
Collapse
Affiliation(s)
| | - Alejandro Cifuentes
- Foodomics Laboratory, CIAL, CSIC-UAM, Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Alberto Valdés
- Foodomics Laboratory, CIAL, CSIC-UAM, Nicolás Cabrera 9, 28049 Madrid, Spain.
| |
Collapse
|
6
|
Blumenstock S, Schulz-Trieglaff EK, Voelkl K, Bolender AL, Lapios P, Lindner J, Hipp MS, Hartl FU, Klein R, Dudanova I. Fluc-EGFP reporter mice reveal differential alterations of neuronal proteostasis in aging and disease. EMBO J 2021; 40:e107260. [PMID: 34410010 PMCID: PMC8488555 DOI: 10.15252/embj.2020107260] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022] Open
Abstract
The cellular protein quality control machinery is important for preventing protein misfolding and aggregation. Declining protein homeostasis (proteostasis) is believed to play a crucial role in age‐related neurodegenerative disorders. However, how neuronal proteostasis capacity changes in different diseases is not yet sufficiently understood, and progress in this area has been hampered by the lack of tools to monitor proteostasis in mammalian models. Here, we have developed reporter mice for in vivo analysis of neuronal proteostasis. The mice express EGFP‐fused firefly luciferase (Fluc‐EGFP), a conformationally unstable protein that requires chaperones for proper folding, and that reacts to proteotoxic stress by formation of intracellular Fluc‐EGFP foci and by reduced luciferase activity. Using these mice, we provide evidence for proteostasis decline in the aging brain. Moreover, we find a marked reaction of the Fluc‐EGFP sensor in a mouse model of tauopathy, but not in mouse models of Huntington’s disease. Mechanistic investigations in primary neuronal cultures demonstrate that different types of protein aggregates have distinct effects on the cellular protein quality control. Thus, Fluc‐EGFP reporter mice enable new insights into proteostasis alterations in different diseases.
Collapse
Affiliation(s)
- Sonja Blumenstock
- Department of Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | | | - Kerstin Voelkl
- Department of Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Anna-Lena Bolender
- Department of Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Paul Lapios
- Department of Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Jana Lindner
- Department of Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Mark S Hipp
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany.,Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Rüdiger Klein
- Department of Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Irina Dudanova
- Department of Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
7
|
Tangrodchanapong T, Sornkaew N, Yurasakpong L, Niamnont N, Nantasenamat C, Sobhon P, Meemon K. Beneficial Effects of Cyclic Ether 2-Butoxytetrahydrofuran from Sea Cucumber Holothuria scabra against Aβ Aggregate Toxicity in Transgenic Caenorhabditis elegans and Potential Chemical Interaction. Molecules 2021; 26:molecules26082195. [PMID: 33920352 PMCID: PMC8070609 DOI: 10.3390/molecules26082195] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022] Open
Abstract
The pathological finding of amyloid-β (Aβ) aggregates is thought to be a leading cause of untreated Alzheimer’s disease (AD). In this study, we isolated 2-butoxytetrahydrofuran (2-BTHF), a small cyclic ether, from Holothuria scabra and demonstrated its therapeutic potential against AD through the attenuation of Aβ aggregation in a transgenic Caenorhabditis elegans model. Our results revealed that amongst the five H. scabra isolated compounds, 2-BTHF was shown to be the most effective in suppressing worm paralysis caused by Aβ toxicity and in expressing strong neuroprotection in CL4176 and CL2355 strains, respectively. An immunoblot analysis showed that CL4176 and CL2006 treated with 2-BTHF showed no effect on the level of Aβ monomers but significantly reduced the toxic oligomeric form and the amount of 1,4-bis(3-carboxy-hydroxy-phenylethenyl)-benzene (X-34)-positive fibril deposits. This concurrently occurred with a reduction of reactive oxygen species (ROS) in the treated CL4176 worms. Mechanistically, heat shock factor 1 (HSF-1) (at residues histidine 63 (HIS63) and glutamine 72 (GLN72)) was shown to be 2-BTHF’s potential target that might contribute to an increased expression of autophagy-related genes required for the breakdown of the Aβ aggregate, thus attenuating its toxicity. In conclusion, 2-BTHF from H. scabra could protect C. elegans from Aβ toxicity by suppressing its aggregation via an HSF-1-regulated autophagic pathway and has been implicated as a potential drug for AD.
Collapse
Affiliation(s)
- Taweesak Tangrodchanapong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
| | - Nilubon Sornkaew
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bangkok 10140, Thailand; (N.S.); (N.N.)
| | - Laphatrada Yurasakpong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
| | - Nakorn Niamnont
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bangkok 10140, Thailand; (N.S.); (N.N.)
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand;
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
| | - Krai Meemon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
- Correspondence: or ; Tel.: +66-22-015-407
| |
Collapse
|
8
|
Tangrodchanapong T, Sobhon P, Meemon K. Frondoside A Attenuates Amyloid-β Proteotoxicity in Transgenic Caenorhabditis elegans by Suppressing Its Formation. Front Pharmacol 2020; 11:553579. [PMID: 33013392 PMCID: PMC7513805 DOI: 10.3389/fphar.2020.553579] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/24/2020] [Indexed: 11/16/2022] Open
Abstract
Oligomeric assembly of Amyloid-β (Aβ) is the main toxic species that contribute to early cognitive impairment in Alzheimer’s patients. Therefore, drugs that reduce the formation of Aβ oligomers could halt the disease progression. In this study, by using transgenic Caenorhabditis elegans model of Alzheimer’s disease, we investigated the effects of frondoside A, a well-known sea cucumber Cucumaria frondosa saponin with anti-cancer activity, on Aβ aggregation and proteotoxicity. The results showed that frondoside A at a low concentration of 1 µM significantly delayed the worm paralysis caused by Aβ aggregation as compared with control group. In addition, the number of Aβ plaque deposits in transgenic worm tissues was significantly decreased. Frondoside A was more effective in these activities than ginsenoside-Rg3, a comparable ginseng saponin. Immunoblot analysis revealed that the level of small oligomers as well as various high molecular weights of Aβ species in the transgenic C. elegans were significantly reduced upon treatment with frondoside A, whereas the level of Aβ monomers was not altered. This suggested that frondoside A may primarily reduce the level of small oligomeric forms, the most toxic species of Aβ. Frondoside A also protected the worms from oxidative stress and rescued chemotaxis dysfunction in a transgenic strain whose neurons express Aβ. Taken together, these data suggested that low dose of frondoside A could protect against Aβ-induced toxicity by primarily suppressing the formation of Aβ oligomers. Thus, the molecular mechanism of how frondoside A exerts its anti-Aβ aggregation should be studied and elucidated in the future.
Collapse
Affiliation(s)
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Krai Meemon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
9
|
Lim CH, Kaur P, Teo E, Lam VYM, Zhu F, Kibat C, Gruber J, Mathuru AS, Tolwinski NS. Application of optogenetic Amyloid-β distinguishes between metabolic and physical damages in neurodegeneration. eLife 2020; 9:52589. [PMID: 32228858 PMCID: PMC7145416 DOI: 10.7554/elife.52589] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
The brains of Alzheimer’s disease patients show a decrease in brain mass and a preponderance of extracellular Amyloid-β plaques. These plaques are formed by aggregation of polypeptides that are derived from the Amyloid Precursor Protein (APP). Amyloid-β plaques are thought to play either a direct or an indirect role in disease progression, however the exact role of aggregation and plaque formation in the aetiology of Alzheimer’s disease (AD) is subject to debate as the biological effects of soluble and aggregated Amyloid-β peptides are difficult to separate in vivo. To investigate the consequences of formation of Amyloid-β oligomers in living tissues, we developed a fluorescently tagged, optogenetic Amyloid-β peptide that oligomerizes rapidly in the presence of blue light. We applied this system to the crucial question of how intracellular Amyloid-β oligomers underlie the pathologies of A. We use Drosophila, C. elegans and D. rerio to show that, although both expression and induced oligomerization of Amyloid-β were detrimental to lifespan and healthspan, we were able to separate the metabolic and physical damage caused by light-induced Amyloid-β oligomerization from Amyloid-β expression alone. The physical damage caused by Amyloid-β oligomers also recapitulated the catastrophic tissue loss that is a hallmark of late AD. We show that the lifespan deficit induced by Amyloid-β oligomers was reduced with Li+ treatment. Our results present the first model to separate different aspects of disease progression. Alzheimer's disease is a progressive condition that damages the brain over time. The cause is not clear, but a toxic molecule called Amyloid-β peptide seems to play a part. It builds up in the brains of people with Alzheimer's disease, forming hard clumps called plaques. Yet, though the plaques are a hallmark of the disease, experimental treatments designed to break them down do not seem to help. This raises the question – do Amyloid-β plaques actually cause Alzheimer's disease? Answering this question is not easy. One way to study the effect of amyloid plaques is to inject clumps of Amyloid-β peptides into model organisms. This triggers Alzheimer's-like brain damage, but it is not clear why. It remains difficult to tell the difference between the damage caused by the injected Amyloid-β peptides and the damage caused by the solid plaques that they form. For this, researchers need a way to trigger plaque formation directly inside animal brains. This would make it possible to test the effects of plaque-targeting treatments, like the drug lithium. Optogenetics is a technique that uses light to control molecules in living animals. Hsien, Kaur et al. have now used this approach to trigger plaque formation by fusing light-sensitive proteins to Amyloid-β peptides in worms, fruit flies and zebrafish. This meant that the peptides clumped together to form plaques whenever the animals were exposed to blue light. This revealed that, while both the Amyloid-β peptides and the plaques caused damage, the plaques were much more toxic. They damaged cell metabolism and caused tissue loss that resembled late Alzheimer's disease in humans. To find out whether it was possible to test Alzheimer's treatments in these animals, Hsien, Kaur et al. treated them with the drug, lithium. This increased their lifespan, reversing some of the damage caused by the plaques. Alzheimer's disease affects more than 46.8 million people worldwide and is the sixth leading cause of death in the USA. But, despite over 50 years of research, there is no cure. This new plaque-formation technique allows researchers to study the effects of amyloid plaques in living animals, providing a new way to test Alzheimer's treatments. This could be of particular help in studies of experimental drugs that aim to reduce plaque formation.
Collapse
Affiliation(s)
- Chu Hsien Lim
- Science Division, Yale- NUS College, Singapore, Singapore
| | - Prameet Kaur
- Science Division, Yale- NUS College, Singapore, Singapore
| | - Emelyne Teo
- Science Division, Yale- NUS College, Singapore, Singapore
| | | | - Fangchen Zhu
- Science Division, Yale- NUS College, Singapore, Singapore
| | - Caroline Kibat
- Science Division, Yale- NUS College, Singapore, Singapore.,Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore.,Department of Physiology, YLL School of Medicine, Singapore, Singapore
| | - Jan Gruber
- Science Division, Yale- NUS College, Singapore, Singapore.,Department of Biochemistry, National University of Singapore, Singapore, Singapore
| | - Ajay S Mathuru
- Science Division, Yale- NUS College, Singapore, Singapore.,Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore.,Department of Physiology, YLL School of Medicine, Singapore, Singapore
| | | |
Collapse
|
10
|
Machado ML, Arantes LP, da Silveira TL, Zamberlan DC, Cordeiro LM, Obetine FBB, da Silva AF, da Cruz IBM, Soares FAA, Oliveira RDP. Ilex paraguariensis extract provides increased resistance against oxidative stress and protection against Amyloid beta-induced toxicity compared to caffeine in Caenorhabditis elegans. Nutr Neurosci 2019; 24:697-709. [PMID: 31595831 DOI: 10.1080/1028415x.2019.1671694] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ilex paraguariensis is a plant from South America, used to prepare a tea-like beverage rich in caffeine and polyphenols with antioxidant proprieties. Caffeine consumption is associated with a lower risk of age-associated neuropathologies, besides several extracts that have antioxidant proprieties are known to be neuroprotective, and oxidative stress strongly correlates with Aβ-toxicity. This study aims to investigate the neuroprotective effects of the Ilex paraguariensis hydroalcoholic extract (IPHE) and to evaluate if caffeine agent present in IPHE exerts neuroprotective effects in an amyloid beta-peptide (Aβ)-induced toxicity in Caenorhabditis elegans. The wild-type and CL2006 worms were treated with IPHE (2 and 4 mg/mL) or caffeine (200 and 400 μM) since larval stage 1 (L1) until they achieved the required age for each assay. IPHE and caffeine increased the lifespan and appeared to act directly by reactive oxygen species (ROS) scavenger in both wild-type and CL2006 worms, also conferred resistance against oxidative stress in wild-type animals. Furthermore, both treatments delayed Aβ-induced paralysis and decreased AChE activity in CL2006. The protective effect of IPHE against Aβ-induced paralysis was found to be dependent on heat shock factor hsf-1 and FOXO-family transcription factor daf-16, which are respectively involved in aging-related processes and chaperone synthesis, while that of caffeine was dependent only on daf-16. Mechanistically, IPHE and caffeine decreased the levels of Aβ mRNA in the CL2006 worms; however, only IPHE induced expression of the heat shock chaperonin hsp-16.2, involved in protein homeostasis. The results were overall better when treated with IPHE than with caffeine.
Collapse
Affiliation(s)
- Marina Lopes Machado
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Leticia Priscilla Arantes
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Tássia Limana da Silveira
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Daniele Coradini Zamberlan
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Larissa Marafiga Cordeiro
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Fabiane Baptista Bicca Obetine
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Aline Franzen da Silva
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | | | - Felix Alexandre Antunes Soares
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Riva de Paula Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
11
|
Cline EN, Bicca MA, Viola KL, Klein WL. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis 2019; 64:S567-S610. [PMID: 29843241 PMCID: PMC6004937 DOI: 10.3233/jad-179941] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid-β oligomer (AβO) hypothesis was introduced in 1998. It proposed that the brain damage leading to Alzheimer’s disease (AD) was instigated by soluble, ligand-like AβOs. This hypothesis was based on the discovery that fibril-free synthetic preparations of AβOs were potent CNS neurotoxins that rapidly inhibited long-term potentiation and, with time, caused selective nerve cell death (Lambert et al., 1998). The mechanism was attributed to disrupted signaling involving the tyrosine-protein kinase Fyn, mediated by an unknown toxin receptor. Over 4,000 articles concerning AβOs have been published since then, including more than 400 reviews. AβOs have been shown to accumulate in an AD-dependent manner in human and animal model brain tissue and, experimentally, to impair learning and memory and instigate major facets of AD neuropathology, including tau pathology, synapse deterioration and loss, inflammation, and oxidative damage. As reviewed by Hayden and Teplow in 2013, the AβO hypothesis “has all but supplanted the amyloid cascade.” Despite the emerging understanding of the role played by AβOs in AD pathogenesis, AβOs have not yet received the clinical attention given to amyloid plaques, which have been at the core of major attempts at therapeutics and diagnostics but are no longer regarded as the most pathogenic form of Aβ. However, if the momentum of AβO research continues, particularly efforts to elucidate key aspects of structure, a clear path to a successful disease modifying therapy can be envisioned. Ensuring that lessons learned from recent, late-stage clinical failures are applied appropriately throughout therapeutic development will further enable the likelihood of a successful therapy in the near-term.
Collapse
Affiliation(s)
- Erika N Cline
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Maíra Assunção Bicca
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
12
|
Chaari A. Molecular chaperones biochemistry and role in neurodegenerative diseases. Int J Biol Macromol 2019; 131:396-411. [DOI: 10.1016/j.ijbiomac.2019.02.148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023]
|
13
|
Zhu Z, Yang T, Zhang L, Liu L, Yin E, Zhang C, Guo Z, Xu C, Wang X. Inhibiting Aβ toxicity in Alzheimer's disease by a pyridine amine derivative. Eur J Med Chem 2019; 168:330-339. [DOI: 10.1016/j.ejmech.2019.02.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/07/2019] [Accepted: 02/17/2019] [Indexed: 12/13/2022]
|
14
|
Ai L, Yang F, Song J, Chen Y, Xiao L, Wang Q, Wang L, Li H, Lei T, Huang Z. Inhibition of Abeta Proteotoxicity by Paeoniflorin in Caenorhabditis elegans Through Regulation of Oxidative and Heat Shock Stress Responses. Rejuvenation Res 2018; 21:304-312. [DOI: 10.1089/rej.2017.1966] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Liping Ai
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Fan Yang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Institute of Forensic Science, Ministry of Public Security, Beijing, China
| | - Jie Song
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Center for Bioresources & Drug Discovery and School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Chen
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Qiangqiang Wang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Liangyi Wang
- Center for Bioresources & Drug Discovery and School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haifeng Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Center for Bioresources & Drug Discovery and School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tao Lei
- Department of Rehabilitation Medicine, Wuhan Children's Hospital, Wuhan, China
| | - Zebo Huang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Center for Bioresources & Drug Discovery and School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
15
|
Guarana ( Paullinia cupana) Extract Protects Caenorhabditis elegans Models for Alzheimer Disease and Huntington Disease through Activation of Antioxidant and Protein Degradation Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9241308. [PMID: 30116496 PMCID: PMC6079341 DOI: 10.1155/2018/9241308] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/03/2018] [Indexed: 11/20/2022]
Abstract
Guarana (Paullinia cupana) is largely consumed in Brazil in high energy drinks and dietary supplements because of its stimulant activity on the central nervous system. Although previous studies have indicated that guarana has some protective effects in Parkinson's (PD), Alzheimer's (AD), and Huntington's (HD) disease models, the underlying mechanisms are unknown. Here, we investigated the protective effects of guarana hydroalcoholic extract (GHE) in Caenorhabditis elegans models of HD and AD. GHE reduced polyglutamine (polyQ) protein aggregation in the muscle and also reduced polyQ-mediated neuronal death in ASH sensory neurons and delayed β-amyloid-induced paralysis in a caffeine-independent manner. Moreover, GHE's protective effects were not mediated by caloric restriction, antimicrobial effects, or development and reproduction impairment. Inactivation of the transcription factors SKN-1 and DAF-16 by RNAi partially blocked the protective effects of GHE treatment in the AD model. We show that the protective effect of GHE is associated with antioxidant activity and modulation of proteostasis, since it increased the lifespan and proteasome activity, reduced intracellular ROS and the accumulation of autophagosomes, and increased the expression of SOD-3 and HSP-16.2. Our findings suggest that GHE has therapeutic potential in combating age-related diseases associated with protein misfolding and accumulation.
Collapse
|
16
|
Haseeb K, Wang J, Xiao K, Yang KL, Sun PP, Wu XT, Luo Y, Song H, Liu HZ, Zhong JM, Peng KM. Effects of Boron Supplementation on Expression of Hsp70 in the Spleen of African Ostrich. Biol Trace Elem Res 2018; 182:317-327. [PMID: 28730576 DOI: 10.1007/s12011-017-1087-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/23/2017] [Indexed: 11/11/2022]
Abstract
Increased synthesis of heat shock protein 70 (Hsp70) occurs in prokaryotes and eukaryotes in response to physiological, environmental, and chemical exposures, thus allowing the cell survival from fatal conditions. Hsp70 cytoprotective properties may be clarified by its anti-apoptotic function. Boron has been reported to play an essential role in various organ developments and metabolisms. However, it is not known if boron is also able to modulate the Hsp70. In the present study, the actions of boron on ostrich spleen and expression level of Hsp70 were investigated. Thirty healthy ostrich chicks were randomly assigned to six groups: groups I, II, III, IV, V, and VI and fed the basal diet spiked with 0-, 40-, 80-, 160-, 320-, and 640-mg boric acid (BA)/L, respectively, in drinking water. The histomorphological examination in the spleen was done by hematoxylin and eosin (HE) staining. The expression level of Hsp70 was analyzed by immunohistochemistry (IHC) and western blotting, and mRNA expression of Hsp70 was investigated by quantitative real-time PCR (qPCR). In order to investigate apoptosis, TUNEL assay reaction in all treatment groups was analyzed. Our results showed that the histological structure of spleen up to 160 mg/L BA supplementation groups well developed. The Hsp70 expression level first induced at low-dose groups (up to group IV) and then inhibited dramatically in high-dose groups (V and VI) while comparing with the group I (0 mg BA). The TUNEL assay reaction revealed that the cell apoptosis amount was decreased in group IV, but in group V and especially in group VI, it was significantly increased (P < 0.01). Taken altogether, proper dietary boron treatment might stimulate ostrich chick spleen development by promoting the Hsp70 expression level and inhibiting apoptosis, while a high amount of boron supplementation would impair the ostrich spleen structure by inhibiting Hsp70 expression level and promoting cell apoptosis.
Collapse
Affiliation(s)
- Khaliq Haseeb
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Jing Wang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Ke Xiao
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Ke-Li Yang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Peng-Peng Sun
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Xing-Tong Wu
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - You Luo
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Hui Song
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Hua-Zhen Liu
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Ju-Ming Zhong
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Ke-Mei Peng
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
17
|
Müller WEG, Wang S, Ackermann M, Neufurth M, Steffen R, Mecja E, Muñoz-Espí R, Feng Q, Schröder HC, Wang X. Rebalancing β-Amyloid-Induced Decrease of ATP Level by Amorphous Nano/Micro Polyphosphate: Suppression of the Neurotoxic Effect of Amyloid β-Protein Fragment 25-35. Int J Mol Sci 2017; 18:2154. [PMID: 29035351 PMCID: PMC5666835 DOI: 10.3390/ijms18102154] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/11/2017] [Accepted: 10/14/2017] [Indexed: 01/24/2023] Open
Abstract
Morbus Alzheimer neuropathology is characterized by an impaired energy homeostasis of brain tissue. We present an approach towards a potential therapy of Alzheimer disease based on the high-energy polymer inorganic polyphosphate (polyP), which physiologically occurs both in the extracellular and in the intracellular space. Rat pheochromocytoma (PC) 12 cells, as well as rat primary cortical neurons were exposed to the Alzheimer peptide Aβ25-35. They were incubated in vitro with polyphosphate (polyP); ortho-phosphate was used as a control. The polymer remained as Na⁺ salt; or complexed in a stoichiometric ratio to Ca2+ (Na-polyP[Ca2+]); or was processed as amorphous Ca-polyP microparticles (Ca-polyP-MP). Ortho-phosphate was fabricated as crystalline Ca-phosphate nanoparticles (Ca-phosphate-NP). We show that the pre-incubation of PC12 cells and primary cortical neurons with polyP protects the cells against the neurotoxic effect of the Alzheimer peptide Aβ25-35. The strongest effect was observed with amorphous polyP microparticles (Ca-polyP-MP). The effect of the soluble sodium salt; Na-polyP (Na-polyP[Ca2+]) was lower; while crystalline orthophosphate nanoparticles (Ca-phosphate-NP) were ineffective. Ca-polyP-MP microparticles and Na-polyP[Ca2+] were found to markedly enhance the intracellular ATP level. Pre-incubation of Aβ25-35 during aggregate formation, with the polyP preparation before exposure of the cells, had a small effect on neurotoxicity. We conclude that recovery of the compromised energy status in neuronal cells by administration of nontoxic biodegradable Ca-salts of polyP reverse the β-amyloid-induced decrease of adenosine triphosphate (ATP) level. This study contributes to a new routes for a potential therapeutic intervention in Alzheimer's disease pathophysiology.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Shunfeng Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University, Johann Joachim Becher Weg 13, D-55099 Mainz, Germany.
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Renate Steffen
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Egherta Mecja
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Rafael Muñoz-Espí
- Institute of Materials Science (ICMUV), Universitat de València, C/Catedràtic José Beltrán 2, 46980 Paterna, València, Spain.
| | - Qingling Feng
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China.
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| |
Collapse
|
18
|
Sharma N, Khurana N, Muthuraman A. Lower vertebrate and invertebrate models of Alzheimer's disease - A review. Eur J Pharmacol 2017; 815:312-323. [PMID: 28943103 DOI: 10.1016/j.ejphar.2017.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 08/20/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease is a common neurodegenerative disorder which is characterized by the presence of beta- amyloid protein and neurofibrillary tangles (NFTs) in the brain. Till now, various higher vertebrate models have been in use to study the pathophysiology of this disease. But, these models possess some limitations like ethical restrictions, high cost, difficult maintenance of large quantity and lesser reproducibility. Besides, various lower chordate animals like Danio rerio, Drosophila melanogaster, Caenorhabditis elegans and Ciona intestinalis have been proved to be an important model for the in vivo determination of targets of drugs with least limitations. In this article, we reviewed different studies conducted on theses models for the better understanding of the pathophysiology of AD and their subsequent application as a potential tool in the preclinical evaluation of new drugs.
Collapse
Affiliation(s)
- Neha Sharma
- Department of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Navneet Khurana
- Department of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Arunachalam Muthuraman
- Department of Pharmacology, Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur, Punjab, India; Department of Pharmacology, JSS College of Pharmacy, Jagadguru Sri Shivarathreeshwara University, Mysuru 570015, Karnataka, India.
| |
Collapse
|
19
|
Romeo M, Stravalaci M, Beeg M, Rossi A, Fiordaliso F, Corbelli A, Salmona M, Gobbi M, Cagnotto A, Diomede L. Humanin Specifically Interacts with Amyloid-β Oligomers and Counteracts Their in vivo Toxicity. J Alzheimers Dis 2017; 57:857-871. [PMID: 28282805 DOI: 10.3233/jad-160951] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The 24-residue peptide humanin (HN) has been proposed as a peptide-based inhibitor able to interact directly with amyloid-β (Aβ) oligomers and interfere with the formation and/or biological properties of toxic Aβ species. When administered exogenously, HN, or its synthetic S14G-derivative (HNG), exerted multiple cytoprotective effects, counteracting the Aβ-induced toxicity. Whether these peptides interact directly with Aβ, particularly with the soluble oligomeric assemblies, remains largely unknown. We here investigated the ability of HN and HNG to interact directly with highly aggregating Aβ42, and interfere with the formation and toxicity of its oligomers. Experiments were run in cell-free conditions and in vivo in a transgenic C. elegans strain in which the Aβ toxicity was specifically due to oligomeric species. Thioflavin-T assay indicated that both HN and HNG delay the formation and reduce the final amount of Aβ42 fibrils. In vitro surface plasmon resonance studies indicated that they interact with Aβ42 oligomers favoring the formation of amorphous larger assemblies, observed with turbidity and electron microscopy. In vivo studies indicated that both HN and HNG decrease the relative abundance of A11-positive prefibrillar oligomers as well as OC-positive fibrillar oligomers and had similar protective effects. However, while HN possibly decreased the oligomers by promoting their assembly into larger aggregates, the reduction of oligomers caused by HNG can be ascribed to a marked decrease of the total Aβ levels, likely the consequence of the HNG-induced overexpression of the Aβ-degrading enzyme neprilysin. These findings provide information on the mechanisms underlying the anti-oligomeric effects of HN and HNG and illustrate the role of S14G substitution in regulating the in vivo mechanism of action.
Collapse
Affiliation(s)
- Margherita Romeo
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Matteo Stravalaci
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Marten Beeg
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Alessandro Rossi
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Fabio Fiordaliso
- Department of Cardiovascular Research, Unit of Bio-imaging, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Alessandro Corbelli
- Department of Cardiovascular Research, Unit of Bio-imaging, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Marco Gobbi
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Alfredo Cagnotto
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| |
Collapse
|
20
|
Sciacca MFM, Romanucci V, Zarrelli A, Monaco I, Lolicato F, Spinella N, Galati C, Grasso G, D’Urso L, Romeo M, Diomede L, Salmona M, Bongiorno C, Di Fabio G, La Rosa C, Milardi D. Inhibition of Aβ Amyloid Growth and Toxicity by Silybins: The Crucial Role of Stereochemistry. ACS Chem Neurosci 2017; 8:1767-1778. [PMID: 28562008 DOI: 10.1021/acschemneuro.7b00110] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The self-assembling of the amyloid β (Aβ) peptide into neurotoxic aggregates is considered a central event in the pathogenesis of Alzheimer's disease (AD). Based on the "amyloid hypothesis", many efforts have been devoted to designing molecules able to halt disease progression by inhibiting Aβ self-assembly. Here, we combine biophysical (ThT assays, TEM and AFM imaging), biochemical (WB and ESI-MS), and computational (all-atom molecular dynamics) techniques to investigate the capacity of four optically pure components of the natural product silymarin (silybin A, silybin B, 2,3-dehydrosilybin A, 2,3-dehydrosilybin B) to inhibit Aβ aggregation. Despite TEM analysis demonstrated that all the four investigated flavonoids prevent the formation of mature fibrils, ThT assays, WB and AFM investigations showed that only silybin B was able to halt the growth of small-sized protofibrils thus promoting the formation of large, amorphous aggregates. Molecular dynamics (MD) simulations indicated that silybin B interacts mainly with the C-terminal hydrophobic segment 35MVGGVV40 of Aβ40. Consequently to silybin B binding, the peptide conformation remains predominantly unstructured along all the simulations. By contrast, silybin A interacts preferentially with the segments 17LVFF20 and 27NKGAII32 of Aβ40 which shows a high tendency to form bend, turn, and β-sheet conformation in and around these two domains. Both 2,3-dehydrosilybin enantiomers bind preferentially the segment 17LVFF20 but lead to the formation of different small-sized, ThT-positive Aβ aggregates. Finally, in vivo studies in a transgenic Caenorhabditis elegans strain expressing human Aβ indicated that silybin B is the most effective of the four compounds in counteracting Aβ proteotoxicity. This study underscores the pivotal role of stereochemistry in determining the neuroprotective potential of silybins and points to silybin B as a promising lead compound for further development in anti-AD therapeutics.
Collapse
Affiliation(s)
- Michele. F. M. Sciacca
- Institute of Biostructures and
Bioimages—Catania, National Research Council, Via Paolo Gaifami 8, 95126 Catania, Italy
| | - Valeria Romanucci
- Department of Chemical Sciences, University of Napoli “Federico II”, Via Cintia 4, I-80126 Napoli, Italy
| | - Armando Zarrelli
- Department of Chemical Sciences, University of Napoli “Federico II”, Via Cintia 4, I-80126 Napoli, Italy
| | - Irene Monaco
- Institute of Biostructures and
Bioimages—Catania, National Research Council, Via Paolo Gaifami 8, 95126 Catania, Italy
| | - Fabio Lolicato
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Department of Physics, Tampere University of Technology, P.O.
Box 692, FI-33101 Tampere, Finland
| | | | - Clelia Galati
- STMicroelectronics, Stradale Primosole 50, 95121 Catania, Italy
| | - Giuseppe Grasso
- Dipartimento di Scienze
Chimiche, Università degli Studi di Catania, Viale Andrea
Doria 6, 95125 Catania, Italy
| | - Luisa D’Urso
- Dipartimento di Scienze
Chimiche, Università degli Studi di Catania, Viale Andrea
Doria 6, 95125 Catania, Italy
| | - Margherita Romeo
- IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Via Giuseppe La Masa 19, 20156, Milano, Italy
| | - Luisa Diomede
- IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Via Giuseppe La Masa 19, 20156, Milano, Italy
| | - Mario Salmona
- IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Via Giuseppe La Masa 19, 20156, Milano, Italy
| | - Corrado Bongiorno
- Institute for Microelectronics
and Microsystems, National Research Council, Stradale Primosole 50, 95121 Catania, Italy
| | - Giovanni Di Fabio
- Department of Chemical Sciences, University of Napoli “Federico II”, Via Cintia 4, I-80126 Napoli, Italy
| | - Carmelo La Rosa
- Dipartimento di Scienze
Chimiche, Università degli Studi di Catania, Viale Andrea
Doria 6, 95125 Catania, Italy
| | - Danilo Milardi
- Institute of Biostructures and
Bioimages—Catania, National Research Council, Via Paolo Gaifami 8, 95126 Catania, Italy
| |
Collapse
|
21
|
Zhang Z, Ma H, Wang X, Zhao Z, Zhang Y, Zhao B, Guo Y, Xu L. A tetrapeptide from maize protects a transgenic Caenorhabditis elegans Aβ1-42model from Aβ-induced toxicity. RSC Adv 2016. [DOI: 10.1039/c6ra06130c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A food-derived bioactive peptide that works as an important antioxidantin vivocould be used to remedy oxidative stress-related diseases.
Collapse
Affiliation(s)
- Zhixian Zhang
- Key Laboratory for Molecular Enzymology and Engineering
- The Ministry of Education
- National Engineering Laboratory for AIDS Vaccine
- School of Life Sciences
- Jilin University
| | - Heran Ma
- Key Laboratory for Molecular Enzymology and Engineering
- The Ministry of Education
- National Engineering Laboratory for AIDS Vaccine
- School of Life Sciences
- Jilin University
| | - Xiaoying Wang
- Jilin Province People's Hospital
- Changchun 130021
- China
| | - Ziyuan Zhao
- Key Laboratory for Molecular Enzymology and Engineering
- The Ministry of Education
- National Engineering Laboratory for AIDS Vaccine
- School of Life Sciences
- Jilin University
| | - Yue Zhang
- Key Laboratory for Molecular Enzymology and Engineering
- The Ministry of Education
- National Engineering Laboratory for AIDS Vaccine
- School of Life Sciences
- Jilin University
| | - Baolu Zhao
- State Key Laboratory of Brain and Cognitive Science
- Institute of Biophysics
- Chinese Academy of Sciences
- Beijing 100101
- China
| | - Yi Guo
- Key Laboratory for Molecular Enzymology and Engineering
- The Ministry of Education
- National Engineering Laboratory for AIDS Vaccine
- School of Life Sciences
- Jilin University
| | - Li Xu
- Key Laboratory for Molecular Enzymology and Engineering
- The Ministry of Education
- National Engineering Laboratory for AIDS Vaccine
- School of Life Sciences
- Jilin University
| |
Collapse
|
22
|
A Cultivated Form of a Red Seaweed (Chondrus crispus), Suppresses β-Amyloid-Induced Paralysis in Caenorhabditis elegans. Mar Drugs 2015; 13:6407-24. [PMID: 26492254 PMCID: PMC4626697 DOI: 10.3390/md13106407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/23/2015] [Accepted: 09/30/2015] [Indexed: 11/17/2022] Open
Abstract
We report here the protective effects of a methanol extract from a cultivated strain of the red seaweed, Chondrus crispus, against β-amyloid-induced toxicity, in a transgenic Caenorhabditis elegans, expressing human Aβ1-42 gene. The methanol extract of C. crispus (CCE), delayed β-amyloid-induced paralysis, whereas the water extract (CCW) was not effective. The CCE treatment did not affect the transcript abundance of amy1; however, Western blot analysis revealed a significant decrease of Aβ species, as compared to untreated worms. The transcript abundance of stress response genes; sod3, hsp16.2 and skn1 increased in CCE-treated worms. Bioassay guided fractionation of the CCE yielded a fraction enriched in monogalactosyl diacylglycerols (MGDG) that significantly delayed the onset of β-amyloid-induced paralysis. Taken together, these results suggested that the cultivated strain of C. crispus, whilst providing dietary nutritional value, may also have significant protective effects against β-amyloid-induced toxicity in C. elegans, partly through reduced β-amyloid species, up-regulation of stress induced genes and reduced accumulation of reactive oxygen species (ROS).
Collapse
|
23
|
Smith HL, Li W, Cheetham ME. Molecular chaperones and neuronal proteostasis. Semin Cell Dev Biol 2015; 40:142-52. [PMID: 25770416 PMCID: PMC4471145 DOI: 10.1016/j.semcdb.2015.03.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/13/2022]
Abstract
Protein homeostasis (proteostasis) is essential for maintaining the functionality of the proteome. The disruption of proteostasis, due to genetic mutations or an age-related decline, leads to aberrantly folded proteins that typically lose their function. The accumulation of misfolded and aggregated protein is also cytotoxic and has been implicated in the pathogenesis of neurodegenerative diseases. Neurons have developed an intrinsic protein quality control network, of which molecular chaperones are an essential component. Molecular chaperones function to promote efficient folding and target misfolded proteins for refolding or degradation. Increasing molecular chaperone expression can suppress protein aggregation and toxicity in numerous models of neurodegenerative disease; therefore, molecular chaperones are considered exciting therapeutic targets. Furthermore, mutations in several chaperones cause inherited neurodegenerative diseases. In this review, we focus on the importance of molecular chaperones in neurodegenerative diseases, and discuss the advances in understanding their protective mechanisms.
Collapse
Affiliation(s)
- Heather L Smith
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Wenwen Li
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | |
Collapse
|
24
|
Luo Y, Yue W, Quan X, Wang Y, Zhao B, Lu Z. Asymmetric dimethylarginine exacerbates Aβ-induced toxicity and oxidative stress in human cell and Caenorhabditis elegans models of Alzheimer disease. Free Radic Biol Med 2015; 79:117-26. [PMID: 25499850 DOI: 10.1016/j.freeradbiomed.2014.12.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 12/02/2014] [Accepted: 12/02/2014] [Indexed: 11/30/2022]
Abstract
Growing evidence suggests a strong association between cardiovascular risk factors and incidence of Alzheimer disease (AD). Asymmetric dimethylarginine (ADMA), the endogenous nitric oxide synthase inhibitor, has been identified as an independent cardiovascular risk factor and is also increased in plasma of patients with AD. However, whether ADMA is involved in the pathogenesis of AD is unknown. In this study, we found that ADMA content was increased in a transgenic Caenorhabditis elegans β-amyloid (Aβ) overexpression model, strain CL2006, and in human SH-SY5Y cells overexpressing the Swedish mutant form of human Aβ precursor protein (APPsw). Moreover, ADMA treatment exacerbated Aβ-induced paralysis and oxidative stress in CL2006 worms and further elevated oxidative stress and Aβ secretion in APPsw cells. Knockdown of type 1 protein arginine N-methyltransferase to reduce ADMA production failed to show a protective effect against Aβ toxicity, but resulted in more paralysis in CL2006 worms as well as increased oxidative stress and Aβ secretion in APPsw cells. However, overexpression of dimethylarginine dimethylaminohydrolase 1 (DDAH1) to promote ADMA degradation significantly attenuated oxidative stress and Aβ secretion in APPsw cells. Collectively, our data support the hypothesis that elevated ADMA contributes to the pathogenesis of AD. Our findings suggest that strategies to increase DDAH1 activity in neuronal cells may be a novel approach to attenuating AD development.
Collapse
Affiliation(s)
- Yunfeng Luo
- College of Life Sciences, University of the Chinese Academy of Science, Beijing 100049, China
| | - Wenhui Yue
- College of Life Sciences, University of the Chinese Academy of Science, Beijing 100049, China
| | - Xin Quan
- College of Life Sciences, University of the Chinese Academy of Science, Beijing 100049, China
| | - Yue Wang
- College of Life Sciences, University of the Chinese Academy of Science, Beijing 100049, China
| | - Baolu Zhao
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Zhongbing Lu
- College of Life Sciences, University of the Chinese Academy of Science, Beijing 100049, China.
| |
Collapse
|
25
|
Perez FP, Bose D, Maloney B, Nho K, Shah K, Lahiri DK. Late-onset Alzheimer's disease, heating up and foxed by several proteins: pathomolecular effects of the aging process. J Alzheimers Dis 2014; 40:1-17. [PMID: 24326519 PMCID: PMC4126605 DOI: 10.3233/jad-131544] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common neurodegenerative disorder in older adults, affecting over 50% of those over age 85. Aging is the most important risk factor for the development of LOAD. Aging is associated with the decrease in the ability of cells to cope with cellular stress, especially protein aggregation. Here we describe how the process of aging affects pathways that control the processing and degradation of abnormal proteins including amyloid-β (Aβ). Genetic association studies in LOAD have successfully identified a large number of genetic variants involved in the development of the disease. However, there is a gap in understanding the interconnections between these pathomolecular events that prevent us from discovering therapeutic targets. We propose novel, pertinent links to elucidate how the biology of aging affects the sequence of events in the development of LOAD. Furthermore we analyze and synthesize the molecular-pathologic-clinical correlations of the aging process, involving the HSF1 and FOXO family pathways, Aβ metabolic pathway, and the different clinical stages of LOAD. Our new model postulates that the aging process would precede Aβ accumulation, and attenuation of HSF1 is an "upstream" event in the cascade that results in excess Aβ and synaptic dysfunction, which may lead to cognitive impairment and/or trigger "downstream" neurodegeneration and synaptic loss. Specific host factors, such as the activity of FOXO family pathways, would mediate the response to Aβ toxicity and the pace of progression toward the clinical manifestations of AD.
Collapse
Affiliation(s)
- Felipe P. Perez
- Department of Medicine, Geriatric Medicine Division, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David Bose
- Department of Medicine, Geriatric Medicine Division, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan Maloney
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kavita Shah
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Debomoy K. Lahiri
- Department of Psychiatry, and of Medical & Molecular Genetics, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
26
|
Zhu Y, Lu X, Wu D, Cai S, Li S, Teng X. The effect of manganese-induced cytotoxicity on mRNA expressions of HSP27, HSP40, HSP60, HSP70 and HSP90 in chicken spleen lymphocytes in vitro. Biol Trace Elem Res 2013; 156:144-52. [PMID: 24081778 DOI: 10.1007/s12011-013-9817-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 09/05/2013] [Indexed: 01/10/2023]
Abstract
The purpose of this study was to investigate the effect of manganese (Mn)-induced cytotoxicity on heat shock proteins in chicken spleen lymphocytes. Lymphocytes were cultured in medium in the absence and presence of MnCl2 (2 × 10(-4), 4 × 10(-4), 6 × 10(-4), 8 × 10(-4), 10 × 10(-4), and 12 × 10(-4) mmol/L) for 12, 24, 36, and 48 h in vitro. Then, the mRNA levels of HSP27, HSP40, HSP60, HSP70, and HSP90 were examined by real-time quantitative PCR. The results showed that the mRNA levels of HSP27, HSP40, HSP60, HSP70, and HSP90 in all treatment groups at all time points, except mRNA levels of HSP27 at 48 h, had the same tendency. As manganese concentration increased, the mRNA expression of the heat shock proteins first increased and then decreased. In other words, we demonstrated that the mRNA expression of the heat shock proteins was induced at lower concentrations of manganese and was inhibited at higher concentrations. Mn had a dosage-dependent effect on HSP27, HSP40, HSP60, HSP70, and HSP90 mRNA expression in chicken spleen lymphocytes in vitro.
Collapse
Affiliation(s)
- Yihao Zhu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | | | | | | | | | | |
Collapse
|
27
|
Li J, Le W. Modeling neurodegenerative diseases in Caenorhabditis elegans. Exp Neurol 2013; 250:94-103. [PMID: 24095843 DOI: 10.1016/j.expneurol.2013.09.024] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 09/18/2013] [Accepted: 09/21/2013] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases which include Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington disease (HD), and others are becoming an increasing threat to human health worldwide. The degeneration and death of certain specific groups of neurons are the hallmarks of these diseases. Despite the research progress in identification of several disease-related genes, the mechanisms underlying the neurodegeneration in these diseases remain unclear. Given the molecular conservation in neuronal signaling between Caenorhabditis elegans and vertebrates, an increasing number of research scientists have used the nematode to study this group of diseases. This review paper will focus on the model system that has been established in C. elegans to investigate the pathogenetic roles of those reported disease-related genes in AD, PD, ALS, HD and others. The progress in C. elegans provides useful information of the genetic interactions and molecular pathways that are critical in the disease process, and may help better our understanding of the disease mechanisms and search for new therapeutics for these devastating diseases.
Collapse
Affiliation(s)
- Jia Li
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, P.R. China
| | | |
Collapse
|
28
|
Novel approaches for studying amyloidogenic peptides/proteins. Curr Opin Pharmacol 2013; 13:797-801. [PMID: 23742923 DOI: 10.1016/j.coph.2013.05.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 05/08/2013] [Accepted: 05/08/2013] [Indexed: 12/22/2022]
Abstract
A growing number of important human diseases are associated with the aggregation and deposition of incorrectly folded proteins in the form of highly structured amyloid fibrils. The aggregation process involves the formation of intermediate oligomeric assemblies with toxic properties. There are many commonalities among the different amyloid diseases, that is, similarities in structural features of amyloid fibrils or determinants of oligomer toxicity. Thus, a better knowledge of the general mechanisms of protein aggregation, the characterization of the aggregate's toxicity, and the identification of compounds interfering with these processes, may help for developing therapeutic strategies for different diseases. A variety of analytical methods are currently applied for these purposes. Here we focus on new applications of consolidated technologies which allow one to obtain informative data in a rapid and convenient manner. In particular, we discuss new applications of Surface Plasmon Resonance to study fibril elongation and to specifically recognize oligomers, as well as to screen for ligands of aggregated species and inhibitors of oligomer formation. We also review new advances in the use of wild-type or transgenic Caenorhabditis elegans as suitable in vivo models for the rapid and relatively inexpensive analysis of oligomer toxicity and for testing putative antagonists against this toxicity.
Collapse
|
29
|
Pierce A, Podlutskaya N, Halloran JJ, Hussong SA, Lin PY, Burbank R, Hart MJ, Galvan V. Over-expression of heat shock factor 1 phenocopies the effect of chronic inhibition of TOR by rapamycin and is sufficient to ameliorate Alzheimer's-like deficits in mice modeling the disease. J Neurochem 2013; 124:880-93. [PMID: 23121022 PMCID: PMC6762020 DOI: 10.1111/jnc.12080] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 10/05/2012] [Accepted: 10/10/2012] [Indexed: 01/17/2023]
Abstract
Rapamycin, an inhibitor of target-of-rapamycin, extends lifespan in mice, possibly by delaying aging. We recently showed that rapamycin halts the progression of Alzheimer's (AD)-like deficits, reduces amyloid-beta (Aβ) and induces autophagy in the human amyloid precursor protein (PDAPP) mouse model. To delineate the mechanisms by which chronic rapamycin delays AD we determined proteomic signatures in brains of control- and rapamycin-treated PDAPP mice. Proteins with reported chaperone-like activity were overrepresented among proteins up-regulated in rapamycin-fed PDAPP mice and the master regulator of the heat-shock response, heat-shock factor 1, was activated. This was accompanied by the up-regulation of classical chaperones/heat shock proteins (HSPs) in brains of rapamycin-fed PDAPP mice. The abundance of most HSP mRNAs except for alpha B-crystallin, however, was unchanged, and the cap-dependent translation inhibitor 4E-BP was active, suggesting that increased expression of HSPs and proteins with chaperone activity may result from preferential translation of pre-existing mRNAs as a consequence of inhibition of cap-dependent translation. The effects of rapamycin on the reduction of Aβ, up-regulation of chaperones, and amelioration of AD-like cognitive deficits were recapitulated by transgenic over-expression of heat-shock factor 1 in PDAPP mice. These results suggest that, in addition to inducing autophagy, rapamycin preserves proteostasis by increasing chaperones. We propose that the failure of proteostasis associated with aging may be a key event enabling AD, and that chronic inhibition of target-of-rapamycin may delay AD by maintaining proteostasis in brain. Read the Editorial Highlight for this article on doi: 10.1111/jnc.12098.
Collapse
Affiliation(s)
- Anson Pierce
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Natalia Podlutskaya
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jonathan J. Halloran
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stacy A. Hussong
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Pei-Yi Lin
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Raquel Burbank
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Matthew J. Hart
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Veronica Galvan
- The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
30
|
Sangha JS, Sun X, Wally OSD, Zhang K, Ji X, Wang Z, Wang Y, Zidichouski J, Prithiviraj B, Zhang J. Liuwei Dihuang (LWDH), a traditional Chinese medicinal formula, protects against β-amyloid toxicity in transgenic Caenorhabditis elegans. PLoS One 2012; 7:e43990. [PMID: 22952840 PMCID: PMC3431378 DOI: 10.1371/journal.pone.0043990] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 07/27/2012] [Indexed: 12/12/2022] Open
Abstract
Liuwei Dihuang (LWDH), a classic Chinese medicinal formula, has been used to improve or restore declined functions related to aging and geriatric diseases, such as impaired mobility, vision, hearing, cognition and memory. Here, we report on the effect and possible mechanisms of LWDH mediated protection of β-amyloid (Aβ) induced paralysis in Caenorhabditis elegans using ethanol extract (LWDH-EE) and water extract (LWDH-WE). Chemical profiling and quantitative analysis revealed the presence of different levels of bioactive components in these extracts. LWDH-WE was rich in polar components such as monosaccharide dimers and trimers, whereas LWDH-EE was enriched in terms of phenolic compounds such as gallic acid and paeonol. In vitro studies revealed higher DPPH radical scavenging activity for LWDH-EE as compared to that found for LWDH-WE. Neither LWDH-EE nor LWDH-WE were effective in inhibiting aggregation of Aβ in vitro. By contrast, LWDH-EE effectively delayed Aβ induced paralysis in the transgenic C. elegans (CL4176) model which expresses human Aβ1–42. Western blot revealed no treatment induced reduction in Aβ accumulation in CL4176 although a significant reduction was observed at an early stage with respect to β-amyloid deposition in C. elegans strain CL2006 which constitutively expresses human Aβ1–42. In addition, LWDH-EE reduced in vivo reactive oxygen species (ROS) in C. elegans (CL4176) that correlated with increased survival of LWDH-EE treated N2 worms under juglone-induced oxidative stress. Analysis with GFP reporter strain TJ375 revealed increased expression of hsp16.2::GFP after thermal stress whereas a minute induction was observed for sod3::GFP. Quantitative gene expression analysis revealed that LWDH-EE repressed the expression of amy1 in CL4176 while up-regulating hsp16.2 induced by elevating temperature. Taken together, these results suggest that LWDH extracts, particularly LWDH-EE, alleviated β-amyloid induced toxicity, in part, through up-regulation of heat shock protein, antioxidant activity and reduced ROS in C. elegans.
Collapse
Affiliation(s)
- Jatinder S. Sangha
- Department of Environmental Sciences, Nova Scotia Agricultural College, Truro, Nova Scotia, Canada
| | - Xiaoli Sun
- Institute for Nutrisciences and Health, National Research Council Canada, Charlottetown, Prince Edward Island, Canada
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Owen S. D. Wally
- Department of Environmental Sciences, Nova Scotia Agricultural College, Truro, Nova Scotia, Canada
| | - Kaibin Zhang
- Institute for Nutrisciences and Health, National Research Council Canada, Charlottetown, Prince Edward Island, Canada
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Xiuhong Ji
- Institute for Nutrisciences and Health, National Research Council Canada, Charlottetown, Prince Edward Island, Canada
| | - Zhimin Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yanwen Wang
- Institute for Nutrisciences and Health, National Research Council Canada, Charlottetown, Prince Edward Island, Canada
- Department of Biomedical Sciences, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Jeffrey Zidichouski
- Institute for Nutrisciences and Health, National Research Council Canada, Charlottetown, Prince Edward Island, Canada
| | - Balakrishnan Prithiviraj
- Department of Environmental Sciences, Nova Scotia Agricultural College, Truro, Nova Scotia, Canada
- * E-mail: (BP); (JZ)
| | - Junzeng Zhang
- Institute for Nutrisciences and Health, National Research Council Canada, Charlottetown, Prince Edward Island, Canada
- Department of Chemistry, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
- * E-mail: (BP); (JZ)
| |
Collapse
|
31
|
Gestwicki JE, Garza D. Protein quality control in neurodegenerative disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 107:327-53. [PMID: 22482455 DOI: 10.1016/b978-0-12-385883-2.00003-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The accumulation of misfolded proteins is a common feature of many neurodegenerative diseases. These observations suggest a potential link between these disorders and protein quality control, a collection of cellular pathways that sense damage to proteins and facilitate their turnover. Consistent with this idea, activation of quality control components, such as molecular chaperones, has been shown to be protective in multiple neurodegenerative disease models. In addition, key studies have suggested that quality control deteriorates with age, further supporting a relationship between these processes. In this chapter, we discuss the evidence linking neurodegeneration to quality control and present the emerging models. We also speculate on why proper quality control is so difficult for certain proteins.
Collapse
Affiliation(s)
- Jason E Gestwicki
- Department of Pathology and the Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
32
|
Edwards HV, Cameron RT, Baillie GS. The emerging role of HSP20 as a multifunctional protective agent. Cell Signal 2011; 23:1447-54. [PMID: 21616144 DOI: 10.1016/j.cellsig.2011.05.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 04/20/2011] [Accepted: 05/09/2011] [Indexed: 12/31/2022]
Abstract
The small heat shock proteins (sHSPs) are a highly conserved family of molecular chaperones that are ubiquitously expressed throughout nature. They are transiently upregulated in many tissue types following stressful stimuli. Recently, one member of the sHSP family, HSP20 (HspB6), has been shown to be highly effective as a protective mediator against a number of debilitating pathological conditions, including cardiac hypertrophy and Alzheimer's disease. Hsp20 is also an important modulator of vital physiological processes, such as smooth muscle relaxation and cardiac contractility. This review focuses on the molecular mechanisms employed by HSP20 that allow it to act as an innate protector in the context of cardiovascular and neurological diseases. Emerging evidence for a possible role as an anti-cancer agent is also presented.
Collapse
Affiliation(s)
- H V Edwards
- Molecular Pharmacology Group, Wolfson Link and Davidson Buildings, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | | | | |
Collapse
|
33
|
Roth DM, Balch WE. Modeling general proteostasis: proteome balance in health and disease. Curr Opin Cell Biol 2011; 23:126-34. [PMID: 21131189 PMCID: PMC3077458 DOI: 10.1016/j.ceb.2010.11.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 11/07/2010] [Accepted: 11/08/2010] [Indexed: 12/17/2022]
Abstract
Protein function is generated and maintained by the proteostasis network (PN) (Balch et al. (2008) Science, 319:916). The PN is a modular, yet integrated system unique to each cell type that is sensitive to signaling pathways that direct development and aging, and respond to folding stress. Mismanagement of protein folding and function triggered by genetic, epigenetic and environmental causes poses a major challenge to human health and lifespan. Herein, we address the impact of proteostasis defined by the FoldFx model on our understanding of protein folding and function in biology. FoldFx describes how general proteostasis control (GPC) enables the polypeptide chain sequence to achieve functional balance in the context of the cellular proteome. By linking together the chemical and energetic properties of the protein fold with the composition of the PN we discuss the principle of the proteostasis boundary (PB) as a key component of GPC. The curved surface of the PB observed in 3-dimensional space suggests that the polypeptide chain sequence and the PN operate as an evolutionarily conserved functional unit to generate and sustain protein dynamics required for biology. Modeling general proteostasis provides a rational basis for tackling some of the most challenging diseases facing mankind in the 21st century.
Collapse
Affiliation(s)
- Daniela M. Roth
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037
| | - William E. Balch
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037
- Institute for Childhood and Neglected Diseases, The Scripps Research Institute, La Jolla, California 92037
| |
Collapse
|
34
|
Finka A, Mattoo RUH, Goloubinoff P. Meta-analysis of heat- and chemically upregulated chaperone genes in plant and human cells. Cell Stress Chaperones 2011; 16:15-31. [PMID: 20694844 PMCID: PMC3024091 DOI: 10.1007/s12192-010-0216-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 07/16/2010] [Accepted: 07/19/2010] [Indexed: 12/18/2022] Open
Abstract
Molecular chaperones are central to cellular protein homeostasis. In mammals, protein misfolding diseases and aging cause inflammation and progressive tissue loss, in correlation with the accumulation of toxic protein aggregates and the defective expression of chaperone genes. Bacteria and non-diseased, non-aged eukaryotic cells effectively respond to heat shock by inducing the accumulation of heat-shock proteins (HSPs), many of which molecular chaperones involved in protein homeostasis, in reducing stress damages and promoting cellular recovery and thermotolerance. We performed a meta-analysis of published microarray data and compared expression profiles of HSP genes from mammalian and plant cells in response to heat or isothermal treatments with drugs. The differences and overlaps between HSP and chaperone genes were analyzed, and expression patterns were clustered and organized in a network. HSPs and chaperones only partly overlapped. Heat-shock induced a subset of chaperones primarily targeted to the cytoplasm and organelles but not to the endoplasmic reticulum, which organized into a network with a central core of Hsp90s, Hsp70s, and sHSPs. Heat was best mimicked by isothermal treatments with Hsp90 inhibitors, whereas less toxic drugs, some of which non-steroidal anti-inflammatory drugs, weakly expressed different subsets of Hsp chaperones. This type of analysis may uncover new HSP-inducing drugs to improve protein homeostasis in misfolding and aging diseases.
Collapse
Affiliation(s)
- Andrija Finka
- Department of Plant Molecular Biology, University of Lausanne, 1015 Lausanne, Switzerland
| | - Rayees U. H. Mattoo
- Department of Plant Molecular Biology, University of Lausanne, 1015 Lausanne, Switzerland
| | - Pierre Goloubinoff
- Department of Plant Molecular Biology, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Diomede L, Cassata G, Fiordaliso F, Salio M, Ami D, Natalello A, Doglia SM, De Luigi A, Salmona M. Tetracycline and its analogues protect Caenorhabditis elegans from β amyloid-induced toxicity by targeting oligomers. Neurobiol Dis 2010; 40:424-31. [PMID: 20637283 DOI: 10.1016/j.nbd.2010.07.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/05/2010] [Accepted: 07/07/2010] [Indexed: 02/01/2023] Open
Abstract
The accumulation and deposition of amyloid beta (Aβ) peptide in extracellular dense plaques in the brain is a key phase in Alzheimer's disease (AD). Small oligomeric forms of Aβ are responsible for the toxicity and the early cognitive impairment observed in patients before the amyloid plaque deposits appear. It is essential for the development of an efficient cure for AD to identify compounds that interfere with Aβ aggregation, counteracting the molecular mechanisms involved in conversion of the monomeric amyloid protein into oligomeric and fibrillar forms. Tetracyclines have been proposed for AD therapy, although their effects on the aggregation of Aβ protein, particularly their ability to interact in vivo with the Aβ oligomers and/or aggregates, remain to be understood. Using transgenic Caenorhabditis elegans as a simplified invertebrate model of AD, we evaluated the ability of tetracyclines to interfere with the sequence of events leading to Aβ proteotoxicity. The drugs directly interact with the Aβ assemblies in vivo and reduce Aβ oligomer deposition, protecting C. elegans from oxidative stress and the onset of the paralysis phenotype. These effects were specific, dose-related and not linked to any antibiotic activity, suggesting that the drugs might offer an effective therapeutic strategy to target soluble Aβ aggregates.
Collapse
Affiliation(s)
- Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dimitriadi M, Hart AC. Neurodegenerative disorders: insights from the nematode Caenorhabditis elegans. Neurobiol Dis 2010; 40:4-11. [PMID: 20493260 DOI: 10.1016/j.nbd.2010.05.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 05/06/2010] [Accepted: 05/11/2010] [Indexed: 10/19/2022] Open
Abstract
Neurodegenerative diseases impose a burden on society, yet for the most part, the mechanisms underlying neuronal dysfunction and death in these disorders remain unclear despite the identification of relevant disease genes. Given the molecular conservation in neuronal signaling pathways across vertebrate and invertebrate species, many researchers have turned to the nematode Caenorhabditis elegans to identify the mechanisms underlying neurodegenerative disease pathology. C. elegans can be engineered to express human proteins associated with neurodegeneration; additionally, the function of C. elegans orthologs of human neurodegenerative disease genes can be dissected. Herein, we examine major C. elegans neurodegeneration models that recapitulate many aspects of human neurodegenerative disease and we survey the screens that have identified modifier genes. This review highlights how the C. elegans community has used this versatile organism to model several aspects of human neurodegeneration and how these studies have contributed to our understanding of human disease.
Collapse
Affiliation(s)
- Maria Dimitriadi
- Department of Neuroscience, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | | |
Collapse
|
37
|
Yu YB, Dosanjh L, Lao L, Tan M, Shim BS, Luo Y. Cinnamomum cassia bark in two herbal formulas increases life span in Caenorhabditis elegans via insulin signaling and stress response pathways. PLoS One 2010; 5:e9339. [PMID: 20179756 PMCID: PMC2825258 DOI: 10.1371/journal.pone.0009339] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 01/26/2010] [Indexed: 11/20/2022] Open
Abstract
Background Proving the efficacy and corresponding mode of action of herbal supplements is a difficult challenge for evidence-based herbal therapy. A major hurdle is the complexity of herbal preparations, many of which combine multiple herbs, particularly when the combination is assumed to be vitally important to the effectiveness of the herbal therapy. This issue may be addressed through the use of contemporary methodology and validated animal models. Methods and Principal Findings In this study, two commonly used traditional herbal formulas, Shi Quan Da Bu Tang (SQDB) and Huo Luo Xiao Ling Dan (HLXL) were evaluated using a survival assay and oxidative stress biomarkers in a well-established C. elegans model of aging. HLXL is an eleven herb formula modified from a top-selling traditional herbal formula for the treatment of arthritic joint pain. SQDB consists of ten herbs often used for fatigue and energy, particularly in the aged. We demonstrate here that SQDB significantly extend life span in a C. elegans model of aging. Among all individual herbs tested, two herbs Cinnamomum cassia bark (Chinese pharmaceutical name: Cinnamomi Cortex, CIN) and Panax ginseng root (Chinese pharmaceutical name: Ginseng Radix, GS) significantly extended life span in C. elegans. CIN in both SQDB and HLXL formula extended life span via modulation of multiple longevity assurance genes, including genes involved in insulin signaling and stress response pathways. All the life-span-extending herbs (SQDB, CIN and GS) also attenuated levels of H2O2 and enhanced small heat shock protein expression. Furthermore, the life span-extending herbs significantly delayed human amyloid beta (Aβ)-induced toxicity in transgenic C. elegans expressing human Aβ. Conclusion/Significance These results validate an invertebrate model for rapid, systematic evaluation of commonly used Chinese herbal formulations and may provide insight for designing future evidence-based herbal therapy(s).
Collapse
Affiliation(s)
- Young-Beob Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, United States of America
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
AbstractThe nematode Caenorhabditis elegans is a genetic model organism and the only animal with a complete nervous system wiring diagram. With only 302 neurons and 95 striated muscle cells, a rich array of mutants with defective locomotion and the facility for individual targeted gene knockdown by RNA interference, it lends itself to the exploration of gene function at nerve muscle junctions. With approximately 60% of human disease genes having a C. elegans homologue, there is growing interest in the deployment of lowcost, high-throughput, drug screens of nematode transgenic and mutant strains mimicking aspects of the pathology of devastating human neuromuscular disorders. Here we explore the contributions already made by C. elegans to our understanding of muscular dystrophies (Duchenne and Becker), spinal muscular atrophy, amyotrophic lateral sclerosis, Friedreich’s ataxia, inclusion body myositis and the prospects for contributions to other neuromuscular disorders. A bottleneck to low-cost, in vivo, large-scale chemical library screening for new candidate therapies has been rapid, automated, behavioural phenotyping. Recent progress in quantifying simple swimming (thrashing) movements is making such screening possible and is expediting the translation of drug candidates towards the clinic.
Collapse
|
39
|
The role of molecular chaperones in human misfolding diseases. FEBS Lett 2009; 583:2647-53. [DOI: 10.1016/j.febslet.2009.04.029] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 04/17/2009] [Indexed: 11/23/2022]
|