1
|
Fani G, Coppi E, Errico S, Cherchi F, Gennari M, Barbut D, Vendruscolo M, Zasloff M, Pugliese AM, Chiti F. Natural aminosterols inhibit NMDA receptors with low nanomolar potency. FEBS J 2025. [PMID: 40123295 DOI: 10.1111/febs.70072] [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: 08/23/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 03/25/2025]
Abstract
Abnormal functions of N-methyl-D-aspartate receptors (NMDARs) are associated with many brain disorders, making them primary targets for drug discovery. We show that natural aminosterols inhibit the NMDAR-mediated increase of intracellular calcium ions in cultured primary neurons and neuroblastoma cells. Structural comparison with known NMDAR-negative allosteric modulators, such as pregnanolone-sulfate-2 (PAS), raises the hypothesis that aminosterols have the same mechanism of action. Fluorescence resonance energy transfer (FRET) measurements using labeled NMDAR and the labeled aminosterol trodusquemine (TRO) indicate close spatial proximity, likely arising from binding. Other indirect yet plausible mechanisms for NMDAR inhibition by TRO were excluded. Electrophysiological patch clamp measurements on primary neurons indicate that pre-incubated TRO inhibits NMDA-induced ion currents with a IC50 of 5 nm. Inhibition is observed only after cell membrane pre-adsorption, indicating accessibility to NMDAR from the cell membrane and binding to the transmembrane domains (TMDs) and TMD-ligand-binding domain (LBD) linkers, similarly to PAS. The TRO IC50 is 5000-fold higher than that of PAS and 20-16 000 times higher than those of other inhibitors binding to TMD/TMD-LBD regions, identifying aminosterols as promising and potent NMDAR modulators.
Collapse
Affiliation(s)
- Giulia Fani
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Italy
| | - Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Italy
| | - Silvia Errico
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Italy
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, UK
| | - Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Italy
| | - Martina Gennari
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Italy
| | - Denise Barbut
- Enterin Research Institute Inc., Philadelphia, PA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, UK
| | - Michael Zasloff
- Enterin Research Institute Inc., Philadelphia, PA, USA
- MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, DC, USA
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Italy
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Italy
| |
Collapse
|
2
|
Franklin Z, Hull C, Delibegovic M, Platt B. Pharmacological PTP1B inhibition rescues motor learning, neuroinflammation, and hyperglycaemia in a mouse model of Alzheimer's disease. Exp Neurol 2025; 385:115115. [PMID: 39672227 DOI: 10.1016/j.expneurol.2024.115115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Patients with Alzheimer's Disease (AD) frequently suffer from comorbidities such as type 2 diabetes mellitus (T2DM), accompanied by shared common pathologies such as increased inflammation and impaired glucose homeostasis. Beta-secretase 1 (BACE1), the rate limiting enzyme in AD associated beta-amyloid (Aβ) production, is also implicated in metabolic dysfunction and can increase central and peripheral protein levels of protein tyrosine phosphatase 1B (PTP1B). PTP1B is a validated target in diabetes and obesity, and is a neuroinflammatory regulator involved in degenerative processes. This study investigated the effects of the PTP1B inhibitor, trodusquemine (MSI-1436) on the cognitive and metabolic phenotypes of the neuronal human BACE1 knock-in (PLB4) mouse, a co-morbidity model of AD and T2DM, and their wild-type (PLBWT) controls. METHODS Five-month-old male PLB4 and PLBWT mice received PTP1B inhibitor treatment (1 mg/kg intraperitoneal injection; 5 weeks). Activity and spatial habituation (Phenotyper), motor learning (RotaRod), glucose tolerance, and brain and liver molecular analyses were analysed following treatment. RESULTS Inhibition of PTP1B improved motor learning alongside glucose tolerance in PLB4 mice, without affecting body weight/adiposity. MSI-1436 treatment led to lower protein levels of amyloid precursor protein (APP), reduced astrogliosis and restoration of the endoplasmic chaperone immunoglobulin heavy chain binding protein (BIP) in the brain, alongside decreased insulin receptor substrate-1 (IRS1) and dipeptidyl peptidase-4 (DPP4) proteins in the liver. CONCLUSION We provide evidence that neuronal BACE1 contributes to neuroinflammation and hyperglycaemia in PLB4 mice, and this can be partially rescued by PTP1B inhibition. Targeting PTP1B may therefore offer an attractive therapeutic approach to ameliorate co-morbidity associated pathologies in AD and T2DM.
Collapse
Affiliation(s)
- Zara Franklin
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| | - Claire Hull
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Mirela Delibegovic
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK; Aberdeen Cardiovascular and Diabetes Centre, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Bettina Platt
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
3
|
Barbut D, Perni M, Zasloff M. Anti-aging properties of the aminosterols of the dogfish shark. NPJ AGING 2024; 10:62. [PMID: 39702521 DOI: 10.1038/s41514-024-00188-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Abstract
The development of anti-aging drugs is challenged by both the apparent complexity of the physiological mechanisms involved in aging and the likelihood that many of these mechanisms remain unknown. As a consequence, the development of anti-aging compounds based on the rational targeting of specific pathways has fallen short of the goal. To date, the most impressive compound is rapamycin, a natural bacterial product initially identified as an antifungal, and only subsequently discovered to have anti-aging properties. In this review, we focus on two aminosterols from the dogfish shark, Squalus acanthias, that we discovered initially as broad-spectrum anti-microbial agents. This review is the first to gather together published studies conducted both in vitro and in numerous vertebrate species to demonstrate that these compounds target aging pathways at the cellular level and provide benefits in multiple aging-associated conditions in relevant animal models and in humans. The dogfish aminosterols should be recognized as potential anti-aging drugs.
Collapse
Affiliation(s)
- Denise Barbut
- BAZ Therapeutics, Inc., Philadelphia, PA, 19103, USA
| | - Michele Perni
- BAZ Therapeutics, Inc., Philadelphia, PA, 19103, USA
| | - Michael Zasloff
- BAZ Therapeutics, Inc., Philadelphia, PA, 19103, USA.
- MedStar Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, DC, 20010, USA.
| |
Collapse
|
4
|
Seychell RM, El Saghir A, Vassallo N. Modulation of Biological Membranes Using Small-Molecule Compounds to Counter Toxicity Caused by Amyloidogenic Proteins. MEMBRANES 2024; 14:231. [PMID: 39590617 PMCID: PMC11596372 DOI: 10.3390/membranes14110231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
The transition of peptides or proteins along a misfolding continuum from soluble functional states to pathological aggregates, to ultimately deposit as amyloid fibrils, is a process that underlies an expanding group of human diseases-collectively known as protein-misfolding disorders (PMDs). These include common and debilitating conditions, such as Alzheimer's disease, Parkinson's disease, and type-2 diabetes. Compelling evidence has emerged that the complex interplay between the misfolded proteins and biological membranes is a key determinant of the pathogenic mechanisms by which harmful amyloid entities are formed and exert their cytotoxicity. Most efforts thus far to develop disease-modifying treatments for PMDs have largely focused on anti-aggregation strategies: to neutralise, or prevent the formation of, toxic amyloid species. Herein, we review the critical role of the phospholipid membrane in mediating and enabling amyloid pathogenicity. We consequently propose that the development of small molecules, which have the potential to uniquely modify the physicochemical properties of the membrane and make it more resilient against damage by misfolded proteins, could provide a novel therapeutic approach in PMDs. By way of an example, natural compounds shown to intercalate into lipid bilayers and inhibit amyloid-lipid interactions, such as the aminosterols, squalamine and trodusquamine, cholesterol, ubiquinone, and select polyphenols, are discussed. Such a strategy would provide a novel approach to counter a wide range of toxic biomolecules implicit in numerous human amyloid pathologies.
Collapse
Affiliation(s)
- Raina Marie Seychell
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Adam El Saghir
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, MSD 2080 Msida, Malta
| |
Collapse
|
5
|
Li B, Dettmer U. Interactions of alpha-synuclein with membranes in Parkinson's disease: Mechanisms and therapeutic strategies. Neurobiol Dis 2024; 201:106646. [PMID: 39181187 PMCID: PMC11760337 DOI: 10.1016/j.nbd.2024.106646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/30/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disease worldwide, is marked by the presence of Lewy bodies and Lewy neurites, neuronal lesions containing large amounts of the synaptic protein alpha-synuclein (αS). While the underlying mechanisms of disease progression in PD remain unclear, increasing evidence supports the importance of interactions between αS and cellular membranes in PD pathology. Therefore, understanding the αS-membrane interplay in health and disease is crucial for the development of therapeutic strategies. In this review, we (1) discuss key scenarios of pathological αS-membrane interactions; (2) present in detail therapeutic strategies explicitly reported to modify αS-membrane interactions; and (3) introduce additional therapeutic strategies that may involve aspects of interfering with αS-membrane interaction. This way, we aim to provide a holistic perspective on this important aspect of disease-modifying strategies for PD and other α-synucleinopathies.
Collapse
Affiliation(s)
- Baoyi Li
- Wycombe Abbey, Buckinghamshire HP11 1PE, UK
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Gonçalves PB, Sodero ACR, Cordeiro Y. Natural products targeting amyloid-β oligomer neurotoxicity in Alzheimer's disease. Eur J Med Chem 2024; 276:116684. [PMID: 39032401 DOI: 10.1016/j.ejmech.2024.116684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) constitutes a major global health issue, characterized by progressive neurodegeneration and cognitive impairment, for which no curative treatment is currently available. Current therapeutic approaches are focused on symptom management, highlighting the critical need for disease-modifying therapy. The hallmark pathology of AD involves the aggregation and accumulation of amyloid-β (Aβ) peptides in the brain. Consequently, drug discovery efforts in recent decades have centered on the Aβ aggregation cascade, which includes the transition of monomeric Aβ peptides into toxic oligomers and, ultimately, mature fibrils. Historically, anti-Aβ strategies focused on the clearance of amyloid fibrils using monoclonal antibodies. However, substantial evidence has highlighted the critical role of Aβ oligomers (AβOs) in AD pathogenesis. Soluble AβOs are now recognized as more toxic than fibrils, directly contributing to synaptic impairment, neuronal damage, and the onset of AD. Targeting AβOs has emerged as a promising therapeutic approach to mitigate cognitive decline in AD. Natural products (NPs) have demonstrated promise against AβO neurotoxicity through various mechanisms, including preventing AβO formation, enhancing clearance mechanisms, or converting AβOs into non-toxic species. Understanding the mechanisms by which anti-AβO NPs operate is useful for developing disease-modifying treatments for AD. In this review, we explore the role of NPs in mitigating AβO neurotoxicity for AD drug discovery, summarizing key evidence from biophysical methods, cellular assays, and animal models. By discussing how NPs modulate AβO neurotoxicity across various experimental systems, we aim to provide valuable insights into novel therapeutic strategies targeting AβOs in AD.
Collapse
Affiliation(s)
| | | | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-900, Brazil
| |
Collapse
|
7
|
Morris OM, Toprakcioglu Z, Röntgen A, Cali M, Knowles TPJ, Vendruscolo M. Aggregation of the amyloid-β peptide (Aβ40) within condensates generated through liquid-liquid phase separation. Sci Rep 2024; 14:22633. [PMID: 39349560 PMCID: PMC11442885 DOI: 10.1038/s41598-024-72265-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/05/2024] [Indexed: 10/02/2024] Open
Abstract
The deposition of the amyloid-β (Aβ) peptide into amyloid fibrils is a hallmark of Alzheimer's disease. Recently, it has been reported that some proteins can aggregate and form amyloids through an intermediate pathway involving a liquid-like condensed phase. These observations prompted us to investigate the phase space of Aβ. We thus explored the ability of Aβ to undergo liquid-liquid phase separation, and the subsequent liquid-to-solid transition that takes place within the resulting condensates. Through the use of microfluidic approaches, we observed that the 40-residue form of Αβ (Αβ40) can undergo liquid-liquid phase separation, and that accessing a liquid-like intermediate state enables Αβ40 to self-assemble and aggregate into amyloid fibrils through this pathway. These results prompt further studies to investigate the possible role of Αβ liquid-liquid phase separation and its subsequent aggregation in the context of Alzheimer's disease and more generally on neurodegenerative processes.
Collapse
Affiliation(s)
- Owen M Morris
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Zenon Toprakcioglu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Alexander Röntgen
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Mariana Cali
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, CB3 OHE, UK
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
8
|
Muscat S, Errico S, Danani A, Chiti F, Grasso G. Leveraging Machine Learning-Guided Molecular Simulations Coupled with Experimental Data to Decipher Membrane Binding Mechanisms of Aminosterols. J Chem Theory Comput 2024. [PMID: 38979909 PMCID: PMC11447954 DOI: 10.1021/acs.jctc.4c00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Understanding the molecular mechanisms of the interactions between specific compounds and cellular membranes is essential for numerous biotechnological applications, including targeted drug delivery, elucidation of the drug mechanism of action, pathogen identification, and novel antibiotic development. However, estimation of the free energy landscape associated with solute binding to realistic biological systems is still a challenging task. In this work, we leverage the Time-lagged Independent Component Analysis (TICA) in combination with neural networks (NN) through the Deep-TICA approach for determining the free energy associated with the membrane insertion processes of two natural aminosterol compounds, trodusquemine (TRO), and squalamine (SQ). These compounds are particularly noteworthy because they interact with the outer layer of neuron membranes, protecting them from the toxic action of misfolded proteins involved in neurodegenerative disorders, in both their monomeric and oligomeric forms. We demonstrate how this strategy could be used to generate an effective collective variable for describing solute absorption in the membrane and for estimating free energy landscape of translocation via on-the-fly probability enhanced sampling (OPES) method. In this context, the computational protocol allowed an exhaustive characterization of the aminosterol entry pathway into a neuron-like lipid bilayer. Furthermore, it provided accurate prediction of membrane binding affinities, in close agreement with the experimental binding data obtained by using fluorescently labeled aminosterols and large unilamellar vesicles (LUVs). The findings contribute significantly to our understanding of aminosterol entry pathways and aminosterol-lipid membrane interactions. Finally, the computational methods deployed in this study further demonstrate considerable potential for investigating membrane binding processes.
Collapse
Affiliation(s)
- Stefano Muscat
- Dalle Molle Institute for Artificial Intelligence IDSIA USI-SUPSI, Via la Santa 1 ,Lugano-Viganello 6962, Switzerland
| | - Silvia Errico
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Andrea Danani
- Dalle Molle Institute for Artificial Intelligence IDSIA USI-SUPSI, Via la Santa 1 ,Lugano-Viganello 6962, Switzerland
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Gianvito Grasso
- Dalle Molle Institute for Artificial Intelligence IDSIA USI-SUPSI, Via la Santa 1 ,Lugano-Viganello 6962, Switzerland
| |
Collapse
|
9
|
Li B, Xiao X, Bi M, Jiao Q, Chen X, Yan C, Du X, Jiang H. Modulating α-synuclein propagation and decomposition: Implications in Parkinson's disease therapy. Ageing Res Rev 2024; 98:102319. [PMID: 38719160 DOI: 10.1016/j.arr.2024.102319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/03/2024] [Accepted: 04/27/2024] [Indexed: 05/14/2024]
Abstract
α-Synuclein (α-Syn) is closely related to the pathogenesis of Parkinson's disease (PD). Under pathological conditions, the conformation of α-syn changes and different forms of α-syn lead to neurotoxicity. According to Braak stages, α-syn can propagate in different brain regions, inducing neurodegeneration and corresponding clinical manifestations through abnormal aggregation of Lewy bodies (LBs) and lewy axons in different types of neurons in PD. So far, PD lacks early diagnosis biomarkers, and treatments are mainly targeted at some clinical symptoms. There is no effective therapy to delay the progression of PD. This review first summarized the role of α-syn in physiological and pathological states, and the relationship between α-syn and PD. Then, we focused on the origin, secretion, aggregation, propagation and degradation of α-syn as well as the important regulatory factors in these processes systematically. Finally, we reviewed some potential drug candidates for alleviating the abnormal aggregation of α-syn in order to provide valuable targets for the treatment of PD to cope with the occurrence and progression of this disease.
Collapse
Affiliation(s)
- Beining Li
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Xue Xiao
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Mingxia Bi
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Qian Jiao
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Xi Chen
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Chunling Yan
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Xixun Du
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China.
| | - Hong Jiang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China; School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China.
| |
Collapse
|
10
|
Xu Y, Filice CT, Leonenko Z. Protective effect of trehalose sugar on amyloid-membrane interactions using BLM electrophysiology. Biophys J 2024; 123:1690-1704. [PMID: 38751113 PMCID: PMC11213996 DOI: 10.1016/j.bpj.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/10/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by dementia and memory loss in the elderly population. The amyloid-β peptide (Aβ) is one of the main pathogenic factors in AD and is known to cause damage to neuronal cellular membranes. There is no cure currently available for AD, and new approaches, including preventive strategies, are highly desirable. In this work, we explore the possibility of protecting neuronal membranes from amyloid-induced damage with naturally existing sugar trehalose. Trehalose has been shown to protect plant cellular membranes in extreme conditions and modify Aβ misfolding. We hypothesize that trehalose can protect the neuronal membrane from amyloid toxicity. In this work, we studied the protective effect of trehalose against Aβ1-42-induced damage in model lipid membranes (DPPC/POPC/cholesterol) using atomic force microscopy and black lipid membrane electrophysiology. Our results demonstrate that Aβ1-42 damaged membranes and led to ionic current leakage across these membranes due to the formation of various defects and pores. The presence of trehalose reduced the ion current across membranes caused by Aβ1-42 peptide damage, thus efficiently protecting the membranes. These findings suggest that the trehalose sugar can potentially be useful in protecting neuronal membranes against amyloid toxicity in AD.
Collapse
Affiliation(s)
- Yue Xu
- Department of Physics & Astronomy, University of Waterloo, Waterloo, ON, Canada
| | - Carina Teresa Filice
- Department of Biology, University of Waterloo, Waterloo, ON, Canada; Waterloo Institute for Nanotechnology, Waterloo, ON, Canada
| | - Zoya Leonenko
- Department of Physics & Astronomy, University of Waterloo, Waterloo, ON, Canada; Department of Biology, University of Waterloo, Waterloo, ON, Canada; Waterloo Institute for Nanotechnology, Waterloo, ON, Canada.
| |
Collapse
|
11
|
Rinauro DJ, Chiti F, Vendruscolo M, Limbocker R. Misfolded protein oligomers: mechanisms of formation, cytotoxic effects, and pharmacological approaches against protein misfolding diseases. Mol Neurodegener 2024; 19:20. [PMID: 38378578 PMCID: PMC10877934 DOI: 10.1186/s13024-023-00651-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/17/2023] [Indexed: 02/22/2024] Open
Abstract
The conversion of native peptides and proteins into amyloid aggregates is a hallmark of over 50 human disorders, including Alzheimer's and Parkinson's diseases. Increasing evidence implicates misfolded protein oligomers produced during the amyloid formation process as the primary cytotoxic agents in many of these devastating conditions. In this review, we analyze the processes by which oligomers are formed, their structures, physicochemical properties, population dynamics, and the mechanisms of their cytotoxicity. We then focus on drug discovery strategies that target the formation of oligomers and their ability to disrupt cell physiology and trigger degenerative processes.
Collapse
Affiliation(s)
- Dillon J Rinauro
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Fabrizio Chiti
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| | - Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY, 10996, USA.
| |
Collapse
|
12
|
McVey Neufeld KA, Mao YK, West CL, Ahn M, Hameed H, Iwashita E, Stanisz AM, Forsythe P, Barbut D, Zasloff M, Kunze WA. Squalamine reverses age-associated changes of firing patterns of myenteric sensory neurons and vagal fibres. Commun Biol 2024; 7:80. [PMID: 38200107 PMCID: PMC10781697 DOI: 10.1038/s42003-023-05623-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 11/21/2023] [Indexed: 01/12/2024] Open
Abstract
Vagus nerve signaling is a key component of the gut-brain axis and regulates diverse physiological processes that decline with age. Gut to brain vagus firing patterns are regulated by myenteric intrinsic primary afferent neuron (IPAN) to vagus neurotransmission. It remains unclear how IPANs or the afferent vagus age functionally. Here we identified a distinct ageing code in gut to brain neurotransmission defined by consistent differences in firing rates, burst durations, interburst and intraburst firing intervals of IPANs and the vagus, when comparing young and aged neurons. The aminosterol squalamine changed aged neurons firing patterns to a young phenotype. In contrast to young neurons, sertraline failed to increase firing rates in the aged vagus whereas squalamine was effective. These results may have implications for improved treatments involving pharmacological and electrical stimulation of the vagus for age-related mood and other disorders. For example, oral squalamine might be substituted for or added to sertraline for the aged.
Collapse
Affiliation(s)
| | - Yu-Kang Mao
- Brain-Body Institute, McMaster University, Hamilton, ON, Canada
| | - Christine L West
- Brain-Body Institute, McMaster University, Hamilton, ON, Canada
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Matthew Ahn
- Brain-Body Institute, McMaster University, Hamilton, ON, Canada
| | - Hashim Hameed
- Brain-Body Institute, McMaster University, Hamilton, ON, Canada
| | - Eiko Iwashita
- Brain-Body Institute, McMaster University, Hamilton, ON, Canada
| | | | - Paul Forsythe
- Department of Medicine, 569 Heritage Medical Research Center, University of Alberta, Edmonton, AB, Canada
| | | | - Michael Zasloff
- Enterin, Inc., Philadelphia, PA, USA.
- MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, DC, USA.
| | - Wolfgang A Kunze
- Brain-Body Institute, McMaster University, Hamilton, ON, Canada.
- Department of Biology, McMaster University, Hamilton, ON, Canada.
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
13
|
Siwecka N, Saramowicz K, Galita G, Rozpędek-Kamińska W, Majsterek I. Inhibition of Protein Aggregation and Endoplasmic Reticulum Stress as a Targeted Therapy for α-Synucleinopathy. Pharmaceutics 2023; 15:2051. [PMID: 37631265 PMCID: PMC10459316 DOI: 10.3390/pharmaceutics15082051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
α-synuclein (α-syn) is an intrinsically disordered protein abundant in the central nervous system. Physiologically, the protein regulates vesicle trafficking and neurotransmitter release in the presynaptic terminals. Pathologies related to misfolding and aggregation of α-syn are referred to as α-synucleinopathies, and they constitute a frequent cause of neurodegeneration. The most common α-synucleinopathy, Parkinson's disease (PD), is caused by abnormal accumulation of α-syn in the dopaminergic neurons of the midbrain. This results in protein overload, activation of endoplasmic reticulum (ER) stress, and, ultimately, neural cell apoptosis and neurodegeneration. To date, the available treatment options for PD are only symptomatic and rely on dopamine replacement therapy or palliative surgery. As the prevalence of PD has skyrocketed in recent years, there is a pending issue for development of new disease-modifying strategies. These include anti-aggregative agents that target α-syn directly (gene therapy, small molecules and immunization), indirectly (modulators of ER stress, oxidative stress and clearance pathways) or combine both actions (natural compounds). Herein, we provide an overview on the characteristic features of the structure and pathogenic mechanisms of α-syn that could be targeted with novel molecular-based therapies.
Collapse
Affiliation(s)
| | | | | | | | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (N.S.); (K.S.); (G.G.); (W.R.-K.)
| |
Collapse
|
14
|
Errico S, Lucchesi G, Odino D, Osman EY, Cascella R, Neri L, Capitini C, Calamai M, Bemporad F, Cecchi C, Kinney WA, Barbut D, Relini A, Canale C, Caminati G, Limbocker R, Vendruscolo M, Zasloff M, Chiti F. Quantitative Attribution of the Protective Effects of Aminosterols against Protein Aggregates to Their Chemical Structures and Ability to Modulate Biological Membranes. J Med Chem 2023. [PMID: 37433124 PMCID: PMC10388293 DOI: 10.1021/acs.jmedchem.3c00182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Natural aminosterols are promising drug candidates against neurodegenerative diseases, like Alzheimer and Parkinson, and one relevant protective mechanism occurs via their binding to biological membranes and displacement or binding inhibition of amyloidogenic proteins and their cytotoxic oligomers. We compared three chemically different aminosterols, finding that they exhibited different (i) binding affinities, (ii) charge neutralizations, (iii) mechanical reinforcements, and (iv) key lipid redistributions within membranes of reconstituted liposomes. They also had different potencies (EC50) in protecting cultured cell membranes against amyloid-β oligomers. A global fitting analysis led to an analytical equation describing quantitatively the protective effects of aminosterols as a function of their concentration and relevant membrane effects. The analysis correlates aminosterol-mediated protection with well-defined chemical moieties, including the polyamine group inducing a partial membrane-neutralizing effect (79 ± 7%) and the cholestane-like tail causing lipid redistribution and bilayer mechanical resistance (21 ± 7%), linking quantitatively their chemistry to their protective effects on biological membranes.
Collapse
Affiliation(s)
- Silvia Errico
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Giacomo Lucchesi
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino 50019, Italy
| | - Davide Odino
- Department of Physics, University of Genoa, Genoa 16146, Italy
| | - Enass Youssef Osman
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, The Arab Republic of Egypt
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Lorenzo Neri
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Claudia Capitini
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino 50019, Italy
- Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy
- National Institute of Optics, National Research Council of Italy (CNR), Florence 50125, Italy
| | - Martino Calamai
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino 50019, Italy
- National Institute of Optics, National Research Council of Italy (CNR), Florence 50125, Italy
| | - Francesco Bemporad
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - William A Kinney
- Enterin Research Institute Inc., Philadelphia, Pennsylvania 19103, United States
| | - Denise Barbut
- Enterin Research Institute Inc., Philadelphia, Pennsylvania 19103, United States
| | - Annalisa Relini
- Department of Physics, University of Genoa, Genoa 16146, Italy
| | - Claudio Canale
- Department of Physics, University of Genoa, Genoa 16146, Italy
| | - Gabriella Caminati
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino 50019, Italy
| | - Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Michael Zasloff
- Enterin Research Institute Inc., Philadelphia, Pennsylvania 19103, United States
- MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, District of Columbia 20007, United States
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| |
Collapse
|
15
|
Viles JH. Imaging Amyloid-β Membrane Interactions: Ion-Channel Pores and Lipid-Bilayer Permeability in Alzheimer's Disease. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 135:e202215785. [PMID: 38515735 PMCID: PMC10952214 DOI: 10.1002/ange.202215785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Indexed: 03/08/2023]
Abstract
The accumulation of the amyloid-β peptides (Aβ) is central to the development of Alzheimer's disease. The mechanism by which Aβ triggers a cascade of events that leads to dementia is a topic of intense investigation. Aβ self-associates into a series of complex assemblies with different structural and biophysical properties. It is the interaction of these oligomeric, protofibril and fibrillar assemblies with lipid membranes, or with membrane receptors, that results in membrane permeability and loss of cellular homeostasis, a key event in Alzheimer's disease pathology. Aβ can have an array of impacts on lipid membranes, reports have included: a carpeting effect; a detergent effect; and Aβ ion-channel pore formation. Recent advances imaging these interactions are providing a clearer picture of Aβ induced membrane disruption. Understanding the relationship between different Aβ structures and membrane permeability will inform therapeutics targeting Aβ cytotoxicity.
Collapse
Affiliation(s)
- John H. Viles
- Department of Biochemistry, SBBS, Queen MaryUniversity of LondonUK
| |
Collapse
|
16
|
Viles JH. Imaging Amyloid-β Membrane Interactions: Ion-Channel Pores and Lipid-Bilayer Permeability in Alzheimer's Disease. Angew Chem Int Ed Engl 2023; 62:e202215785. [PMID: 36876912 PMCID: PMC10953358 DOI: 10.1002/anie.202215785] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
The accumulation of the amyloid-β peptides (Aβ) is central to the development of Alzheimer's disease. The mechanism by which Aβ triggers a cascade of events that leads to dementia is a topic of intense investigation. Aβ self-associates into a series of complex assemblies with different structural and biophysical properties. It is the interaction of these oligomeric, protofibril and fibrillar assemblies with lipid membranes, or with membrane receptors, that results in membrane permeability and loss of cellular homeostasis, a key event in Alzheimer's disease pathology. Aβ can have an array of impacts on lipid membranes, reports have included: a carpeting effect; a detergent effect; and Aβ ion-channel pore formation. Recent advances imaging these interactions are providing a clearer picture of Aβ induced membrane disruption. Understanding the relationship between different Aβ structures and membrane permeability will inform therapeutics targeting Aβ cytotoxicity.
Collapse
Affiliation(s)
- John H. Viles
- Department of Biochemistry, SBBS, Queen MaryUniversity of LondonUK
| |
Collapse
|
17
|
Maccari R, Ottanà R. Can Allostery Be a Key Strategy for Targeting PTP1B in Drug Discovery? A Lesson from Trodusquemine. Int J Mol Sci 2023; 24:ijms24119621. [PMID: 37298571 DOI: 10.3390/ijms24119621] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is an enzyme crucially implicated in aberrations of various signaling pathways that underlie the development of different human pathologies, such as obesity, diabetes, cancer, and neurodegenerative disorders. Its inhibition can prevent these pathogenetic events, thus providing a useful tool for the discovery of novel therapeutic agents. The search for allosteric PTP1B inhibitors can represent a successful strategy to identify drug-like candidates by offering the opportunity to overcome some issues related to catalytic site-directed inhibitors, which have so far hampered the development of drugs targeting this enzyme. In this context, trodusquemine (MSI-1436), a natural aminosterol that acts as a non-competitive PTP1B inhibitor, appears to be a milestone. Initially discovered as a broad-spectrum antimicrobial agent, trodusquemine exhibited a variety of unexpected properties, ranging from antidiabetic and anti-obesity activities to effects useful to counteract cancer and neurodegeneration, which prompted its evaluation in several preclinical and clinical studies. In this review article, we provide an overview of the main findings regarding the activities and therapeutic potential of trodusquemine and their correlation with PTP1B inhibition. We also included some aminosterol analogues and related structure-activity relationships that could be useful for further studies aimed at the discovery of new allosteric PTP1B inhibitors.
Collapse
Affiliation(s)
- Rosanna Maccari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Rosaria Ottanà
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
18
|
Bigi A, Lombardo E, Cascella R, Cecchi C. The Toxicity of Protein Aggregates: New Insights into the Mechanisms. Int J Mol Sci 2023; 24:7974. [PMID: 37175681 PMCID: PMC10178715 DOI: 10.3390/ijms24097974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
The aberrant aggregation of specific peptides and proteins is the common feature of a range of more than 50 human pathologies, collectively referred to as protein misfolding diseases [...].
Collapse
|
19
|
Limbocker R, Cremades N, Cascella R, Tessier PM, Vendruscolo M, Chiti F. Characterization of Pairs of Toxic and Nontoxic Misfolded Protein Oligomers Elucidates the Structural Determinants of Oligomer Toxicity in Protein Misfolding Diseases. Acc Chem Res 2023. [PMID: 37071750 DOI: 10.1021/acs.accounts.3c00045] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
ConspectusThe aberrant misfolding and aggregation of peptides and proteins into amyloid aggregates occurs in over 50 largely incurable protein misfolding diseases. These pathologies include Alzheimer's and Parkinson's diseases, which are global medical emergencies owing to their prevalence in increasingly aging populations worldwide. Although the presence of mature amyloid aggregates is a hallmark of such neurodegenerative diseases, misfolded protein oligomers are increasingly recognized as of central importance in the pathogenesis of many of these maladies. These oligomers are small, diffusible species that can form as intermediates in the amyloid fibril formation process or be released by mature fibrils after they are formed. They have been closely associated with the induction of neuronal dysfunction and cell death. It has proven rather challenging to study these oligomeric species because of their short lifetimes, low concentrations, extensive structural heterogeneity, and challenges associated with producing stable, homogeneous, and reproducible populations. Despite these difficulties, investigators have developed protocols to produce kinetically, chemically, or structurally stabilized homogeneous populations of protein misfolded oligomers from several amyloidogenic peptides and proteins at experimentally ameneable concentrations. Furthermore, procedures have been established to produce morphologically similar but structurally distinct oligomers from the same protein sequence that are either toxic or nontoxic to cells. These tools offer unique opportunities to identify and investigate the structural determinants of oligomer toxicity by a close comparative inspection of their structures and the mechanisms of action through which they cause cell dysfunction.This Account reviews multidisciplinary results, including from our own groups, obtained by combining chemistry, physics, biochemistry, cell biology, and animal models for pairs of toxic and nontoxic oligomers. We describe oligomers comprised of the amyloid-β peptide, which underlie Alzheimer's disease, and α-synuclein, which are associated with Parkinson's disease and other related neurodegenerative pathologies, collectively known as synucleinopathies. Furthermore, we also discuss oligomers formed by the 91-residue N-terminal domain of [NiFe]-hydrogenase maturation factor from E. coli, which we use as a model non-disease-related protein, and by an amyloid stretch of Sup35 prion protein from yeast. These oligomeric pairs have become highly useful experimental tools for studying the molecular determinants of toxicity characteristic of protein misfolding diseases. Key properties have been identified that differentiate toxic from nontoxic oligomers in their ability to induce cellular dysfunction. These characteristics include solvent-exposed hydrophobic regions, interactions with membranes, insertion into lipid bilayers, and disruption of plasma membrane integrity. By using these properties, it has been possible to rationalize in model systems the responses to pairs of toxic and nontoxic oligomers. Collectively, these studies provide guidance for the development of efficacious therapeutic strategies to target rationally the cytotoxicity of misfolded protein oligomers in neurodegenerative conditions.
Collapse
Affiliation(s)
- Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Nunilo Cremades
- Institute for Biocomputation and Physics of Complex Systems (BIFI) and Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza 50009, Spain
| | - Roberta Cascella
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| | - Peter M Tessier
- Departments of Chemical Engineering, Pharmaceutical Sciences, and Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Fabrizio Chiti
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| |
Collapse
|
20
|
Peña-Díaz S, García-Pardo J, Ventura S. Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson's Disease. Pharmaceutics 2023; 15:839. [PMID: 36986700 PMCID: PMC10059018 DOI: 10.3390/pharmaceutics15030839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Parkinson's disease, the second most common neurodegenerative disorder worldwide, is characterized by the accumulation of protein deposits in the dopaminergic neurons. These deposits are primarily composed of aggregated forms of α-Synuclein (α-Syn). Despite the extensive research on this disease, only symptomatic treatments are currently available. However, in recent years, several compounds, mainly of an aromatic character, targeting α-Syn self-assembly and amyloid formation have been identified. These compounds, discovered by different approaches, are chemically diverse and exhibit a plethora of mechanisms of action. This work aims to provide a historical overview of the physiopathology and molecular aspects associated with Parkinson's disease and the current trends in small compound development to target α-Syn aggregation. Although these molecules are still under development, they constitute an important step toward discovering effective anti-aggregational therapies for Parkinson's disease.
Collapse
Affiliation(s)
- Samuel Peña-Díaz
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Javier García-Pardo
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
21
|
Reorganization of the outer layer of a model of the plasma membrane induced by a neuroprotective aminosterol. Colloids Surf B Biointerfaces 2023; 222:113115. [PMID: 36603410 DOI: 10.1016/j.colsurfb.2022.113115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/07/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Trodusquemine is an amphipathic aminosterol that has recently shown therapeutic benefit in neurodegenerative diseases altering the binding of misfolded proteins to the cell membrane. To unravel the underlying mechanism, we studied the interactions between Trodusquemine (TRO) and lipid monolayers simulating the outer layer of the plasma membrane. We selected two different compositions of dioleoylphosphatidylcholine (DOPC), sphingomyelin (SM), cholesterol (Chol) and monosialotetrahexosylganglioside (GM1) lipid mixture mimicking either a lipid-raft containing membrane (Ld+So phases) or a single-phase disordered membrane (Ld phase). Surface pressure-area isotherms and surface compressional modulus-area combined with Brewster Angle Microscopy (BAM) provided the thermodynamic and morphological information on the lipid monolayer in the presence of increasing amounts of TRO in the monolayer. Experiments revealed that TRO forms stable spreading monolayers at the buffer-air interface where it undergoes multiple reversible phase transitions to bi- and tri-layers at the interface. When TRO was spread at the interface with the lipid mixtures, we found that it distributes in the lipid monolayer for both the selected lipid compositions, but a maximum TRO uptake in the rafts-containing monolayer was observed for a Lipid/TRO molar ratio equal to 3:2. Statistical analysis of BAM images revealed that TRO induces a decrease in the size of the condensed domains, an increase in their number and in the thickness mismatch between the Ld and So phase. Experiments and MD simulations converge to indicate that TRO adsorbs preferentially at the border of the So domains. Removal of GM1 from the lipid Ld+So mixture resulted in an even greater TRO-mediated reduction of the size of the So domains suggesting that the presence of GM1 hinders the localization of TRO at the So domains boundaries. Taken together these observations suggest that Trodusquemine influences the organization of lipid rafts within the neuronal membrane in a dose-dependent manner whereas it evenly distributes in disordered expanded phases of the membrane model.
Collapse
|
22
|
Gabriel JM, Tan T, Rinauro DJ, Hsu CM, Buettner CJ, Gilmer M, Kaur A, McKenzie TL, Park M, Cohen S, Errico S, Wright AK, Chiti F, Vendruscolo M, Limbocker R. EGCG inactivates a pore-forming toxin by promoting its oligomerization and decreasing its solvent-exposed hydrophobicity. Chem Biol Interact 2023; 371:110307. [PMID: 36535315 DOI: 10.1016/j.cbi.2022.110307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Natural proteinaceous pore-forming agents can bind and permeabilize cell membranes, leading to ion dyshomeostasis and cell death. In the search for antidotes that can protect cells from peptide toxins, we discovered that the polyphenol epigallocatechin gallate (EGCG) interacts directly with melittin from honeybee venom, resulting in the elimination of its binding to the cell membrane and toxicity by markedly lowering the extent of its solvent-exposed hydrophobicity and promoting its oligomerization into larger species. These physicochemical parameters have also been shown to play a key role in the binding to cells of misfolded protein oligomers in a host of neurodegenerative diseases, where oligomer-membrane binding and associated toxicity have been shown to correlate negatively with oligomer size and positively with solvent-exposed hydrophobicity. For melittin, which is not an amyloid-forming protein and has a very distinct mechanism of toxicity compared to misfolded oligomers, we find that the size-hydrophobicity-toxicity relationship also rationalizes the pharmacological attenuation of melittin toxicity by EGCG. These results highlight the importance of the physicochemical properties of pore forming agents in mediating their interactions with cell membranes and suggest a possible therapeutic approach based on compounds with a similar mechanism of action as EGCG.
Collapse
Affiliation(s)
- Justus M Gabriel
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Thomas Tan
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Dillon J Rinauro
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Claire M Hsu
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Caleb J Buettner
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Marshall Gilmer
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Amrita Kaur
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Tristan L McKenzie
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Martin Park
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Sophie Cohen
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Silvia Errico
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK; Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Aidan K Wright
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA.
| |
Collapse
|
23
|
The Role of α-Synuclein in SNARE-mediated Synaptic Vesicle Fusion. J Mol Biol 2023; 435:167775. [PMID: 35931109 DOI: 10.1016/j.jmb.2022.167775] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023]
Abstract
Neuronal communication depends on exquisitely regulated membrane fusion between synaptic vesicles and presynaptic neurons, which results in neurotransmitter release in precisely timed patterns. Presynaptic dysfunctions are known to occur prior to the onset of neurodegenerative diseases, including Parkinson's disease. Synaptic accumulation of α-synuclein (α-Syn) oligomers has been implicated in the pathway leading to such outcomes. α-Syn oligomers exert aberrant effects on presynaptic fusion machinery through their interactions with synaptic vesicles and proteins. Here, we summarize in vitro bulk and single-vesicle assays for investigating the functions of α-Syn monomers and oligomers in synaptic vesicle fusion and then discuss the current understanding of the roles of α-Syn monomers and oligomers in synaptic vesicle fusion. Finally, we suggest a new therapeutic avenue specifically targeting the mechanisms of α-Syn oligomer toxicity rather than the oligomer itself.
Collapse
|
24
|
Capitini C, Pesce L, Fani G, Mazzamuto G, Genovese M, Franceschini A, Paoli P, Pieraccini G, Zasloff M, Chiti F, Pavone FS, Calamai M. Studying the trafficking of labeled trodusquemine and its application as nerve marker for light-sheet and expansion microscopy. FASEB J 2022; 36:e22655. [PMID: 36421008 PMCID: PMC9827910 DOI: 10.1096/fj.202201276r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022]
Abstract
Trodusquemine is an aminosterol with a variety of biological and pharmacological functions, such as acting as an antimicrobial, stimulating body weight loss and interfering with the toxicity of proteins involved in the development of Alzheimer's and Parkinson's diseases. The mechanisms of interaction of aminosterols with cells are, however, still largely uncharacterized. Here, by using fluorescently labeled trodusquemine (TRO-A594 and TRO-ATTO565), we show that trodusquemine binds initially to the plasma membrane of living cells, that the binding affinity is dependent on cholesterol, and that trodusquemine is then internalized and mainly targeted to lysosomes after internalization. We also found that TRO-A594 is able to strongly and selectively bind to myelinated fibers in fixed mouse brain slices, and that it is a marker compatible with tissue clearing and light-sheet fluorescence microscopy or expansion microscopy. In conclusion, this work contributes to further characterize the biology of aminosterols and provides a new tool for nerve labeling suitable for the most advanced microscopy techniques.
Collapse
Affiliation(s)
- Claudia Capitini
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly
| | - Luca Pesce
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly
| | - Giulia Fani
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | - Giacomo Mazzamuto
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly,National Institute of Optics – National Research Council (CNR‐INO)Sesto FiorentinoItaly
| | - Massimo Genovese
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | - Alessandra Franceschini
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | | | - Michael Zasloff
- Enterin Inc.PhiladelphiaPennsylvaniaUSA,MedStar‐Georgetown Transplant InstituteGeorgetown University School of MedicineWashingtonDCUSA
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | - Francesco S. Pavone
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly,National Institute of Optics – National Research Council (CNR‐INO)Sesto FiorentinoItaly
| | - Martino Calamai
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,National Institute of Optics – National Research Council (CNR‐INO)Sesto FiorentinoItaly
| |
Collapse
|
25
|
Uddin A, Malla JA, Kumar H, Kumari M, Sinha S, Sharma VK, Kumar Y, Talukdar P, Lahiri M, Maiti TK, Hazra P. Development of a Systematic Strategy toward Promotion of α-Synuclein Aggregation Using 2-Hydroxyisophthalamide-Based Systems. Biochemistry 2022; 61:2267-2279. [PMID: 36219819 DOI: 10.1021/acs.biochem.2c00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Establishing a potent scheme against α-synuclein aggregation involved in Parkinson's disease has been evaluated as a promising route to identify compounds that either inhibit or promote the aggregation process of α-synuclein. In the last two decades, this perspective has guided a dramatic increase in the efforts, focused on developing potent drugs either for retardation or promotion of the self-assembly process of α-synuclein. To address this issue, using a chemical kinetics platform, we developed a strategy that enabled a progressively detailed analysis of the molecular events leading to protein aggregation at the microscopic level in the presence of a recently synthesized 2-hydroxyisophthalamide class of small organic molecules based on their binding affinity. Furthermore, qualitatively, we have developed a strategy of disintegration of α-synuclein fibrils in the presence of these organic molecules. Finally, we have shown that these organic molecules effectively suppress the toxicity of α-synuclein oligomers in neuron cells.
Collapse
Affiliation(s)
- Aslam Uddin
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Javid Ahmad Malla
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Harish Kumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru560065, India
| | - Manisha Kumari
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad121001, India
| | - Suman Sinha
- Institute of Pharmaceutical Research, GLA University, Mathura281406, India
| | - Virender Kumar Sharma
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Yashwant Kumar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India.,National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru560065, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Mayurika Lahiri
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Tushar Kanti Maiti
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad121001, India
| | - Partha Hazra
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| |
Collapse
|
26
|
Protein tyrosine phosphatase 1B (PTP1B) as a potential therapeutic target for neurological disorders. Biomed Pharmacother 2022; 155:113709. [PMID: 36126456 DOI: 10.1016/j.biopha.2022.113709] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/23/2022] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a typical member of the PTP family, considered a direct negative regulator of several receptor and receptor-associated tyrosine kinases. This widely localized enzyme has been involved in the pathophysiology of several diseases. More recently, PTP1B has attracted attention in the field of neuroscience, since its activation in brain cells can lead to schizophrenia-like behaviour deficits, anxiety-like effects, neurodegeneration, neuroinflammation and depression. Conversely, PTP1B inhibition has been shown to prevent microglial activation, thus exerting a potent anti-inflammatory effect and has also shown potential to increase the cognitive process through the stimulation of hippocampal insulin, leptin and BDNF/TrkB receptors. Notwithstanding, most research on the clinical efficacy of targeting PTP1B has been developed in the field of obesity and type 2 diabetes mellitus (TD2M). However, despite the link existing between these metabolic alterations and neurodegeneration, no clinical trials assessing the neurological advantages of PTP1B inhibition have been performed yet. Preclinical studies, though, have provided strong evidence that targeting PTP1B could allow to reach different pathophysiological mechanisms at once. herefore, specific interventions or trials should be designed to modulate PTP1B activity in brain, since it is a promising strategy to decelerate or prevent neurodegeneration in aged individuals, among other neurological diseases. The present paper fails to include all neurological conditions in which PTP1B could have a role; instead, it focuses on those which have been related to metabolic alterations and neurodegenerative processes. Moreover, only preclinical data is discussed, since clinical studies on the potential of PTP1B inhibition for treating neurological diseases are still required.
Collapse
|
27
|
Kreiser RP, Wright AK, Sasser LR, Rinauro DJ, Gabriel JM, Hsu CM, Hurtado JA, McKenzie TL, Errico S, Albright JA, Richardson L, Jaffett VA, Riegner DE, Nguyen LT, LeForte K, Zasloff M, Hollows JE, Chiti F, Vendruscolo M, Limbocker R. A Brain-Permeable Aminosterol Regulates Cell Membranes to Mitigate the Toxicity of Diverse Pore-Forming Agents. ACS Chem Neurosci 2022; 13:1219-1231. [PMID: 35404569 PMCID: PMC9026273 DOI: 10.1021/acschemneuro.1c00840] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
![]()
The molecular composition
of the plasma membrane plays a key role
in mediating the susceptibility of cells to perturbations induced
by toxic molecules. The pharmacological regulation of the properties
of the cell membrane has therefore the potential to enhance cellular
resilience to a wide variety of chemical and biological compounds.
In this study, we investigate the ability of claramine, a blood–brain
barrier permeable small molecule in the aminosterol class, to neutralize
the toxicity of acute biological threat agents, including melittin
from honeybee venom and α-hemolysin from Staphylococcus
aureus. Our results show that claramine neutralizes
the toxicity of these pore-forming agents by preventing their interactions
with cell membranes without perturbing their structures in a detectable
manner. We thus demonstrate that the exogenous administration of an
aminosterol can tune the properties of lipid membranes and protect
cells from diverse biotoxins, including not just misfolded protein
oligomers as previously shown but also biological protein-based toxins.
Our results indicate that the investigation of regulators of the physicochemical
properties of cell membranes offers novel opportunities to develop
countermeasures against an extensive set of cytotoxic effects associated
with cell membrane disruption.
Collapse
Affiliation(s)
- Ryan P. Kreiser
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Aidan K. Wright
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Liam R. Sasser
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Dillon J. Rinauro
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Justus M. Gabriel
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Claire M. Hsu
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Jorge A. Hurtado
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Tristan L. McKenzie
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Silvia Errico
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| | - J. Alex Albright
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Lance Richardson
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Victor A. Jaffett
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Dawn E. Riegner
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Lam T. Nguyen
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Kathleen LeForte
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Michael Zasloff
- MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, District of Columbia 20010, United States
| | - Jared E. Hollows
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| |
Collapse
|
28
|
Peña-Díaz S, Pujols J, Vasili E, Pinheiro F, Santos J, Manglano-Artuñedo Z, Outeiro TF, Ventura S. The small aromatic compound SynuClean-D inhibits the aggregation and seeded polymerization of multiple α-synuclein strains. J Biol Chem 2022; 298:101902. [PMID: 35390347 PMCID: PMC9079179 DOI: 10.1016/j.jbc.2022.101902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/25/2022] Open
Abstract
Parkinson’s disease is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra, as well as the accumulation of intraneuronal proteinaceous inclusions known as Lewy bodies and Lewy neurites. The major protein component of Lewy inclusions is the intrinsically disordered protein α-synuclein (α-Syn), which can adopt diverse amyloid structures. Different conformational strains of α-Syn have been proposed to be related to the onset of distinct synucleinopathies; however, how specific amyloid fibrils cause distinctive pathological traits is not clear. Here, we generated three different α-Syn amyloid conformations at different pH and salt concentrations and analyzed the activity of SynuClean-D (SC-D), a small aromatic molecule, on these strains. We show that incubation of α-Syn with SC-D reduced the formation of aggregates and the seeded polymerization of α-Syn in all cases. Moreover, we found that SC-D exhibited a general fibril disaggregation activity. Finally, we demonstrate that treatment with SC-D also reduced strain-specific intracellular accumulation of phosphorylated α-Syn inclusions. Taken together, we conclude that SC-D may be a promising hit compound to inhibit polymorphic α-Syn aggregation.
Collapse
Affiliation(s)
- Samuel Peña-Díaz
- Institut de Biotecnologia i Biomedicina. Universitat Autonoma de Barcelona, Bellaterra, Spain; Departament de Bioquimica i Biologia Molecular. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Jordi Pujols
- Institut de Biotecnologia i Biomedicina. Universitat Autonoma de Barcelona, Bellaterra, Spain; Departament de Bioquimica i Biologia Molecular. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Eftychia Vasili
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany; Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Francisca Pinheiro
- Institut de Biotecnologia i Biomedicina. Universitat Autonoma de Barcelona, Bellaterra, Spain; Departament de Bioquimica i Biologia Molecular. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Jaime Santos
- Institut de Biotecnologia i Biomedicina. Universitat Autonoma de Barcelona, Bellaterra, Spain; Departament de Bioquimica i Biologia Molecular. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Zoe Manglano-Artuñedo
- Institut de Biotecnologia i Biomedicina. Universitat Autonoma de Barcelona, Bellaterra, Spain; Departament de Bioquimica i Biologia Molecular. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany; Max Planck Institute for Experimental Medicine, Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, Newcastle, United Kingdom; Scientific Employee With a Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina. Universitat Autonoma de Barcelona, Bellaterra, Spain; Departament de Bioquimica i Biologia Molecular. Universitat Autonoma de Barcelona, Bellaterra, Spain; ICREA, Passeig Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
29
|
Effects of oligomer toxicity, fibril toxicity and fibril spreading in synucleinopathies. Cell Mol Life Sci 2022; 79:174. [PMID: 35244787 PMCID: PMC8897347 DOI: 10.1007/s00018-022-04166-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 12/18/2022]
Abstract
Protein misfolding is a general hallmark of protein deposition diseases, such as Alzheimer’s disease or Parkinson’s disease, in which different types of aggregated species (oligomers, protofibrils and fibrils) are generated by the cells. Despite widespread interest, the relationship between oligomers and fibrils in the aggregation process and spreading remains elusive. A large variety of experimental evidences supported the idea that soluble oligomeric species of different proteins might be more toxic than the larger fibrillar forms. Furthermore, the lack of correlation between the presence of the typical pathological inclusions and disease sustained this debate. However, recent data show that the β-sheet core of the α-Synuclein (αSyn) fibrils is unable to establish persistent interactions with the lipid bilayers, but they can release oligomeric species responsible for an immediate dysfunction of the recipient neurons. Reversibly, such oligomeric species could also contribute to pathogenesis via neuron-to-neuron spreading by their direct cell-to-cell transfer or by generating new fibrils, following their neuronal uptake. In this Review, we discuss the various mechanisms of cellular dysfunction caused by αSyn, including oligomer toxicity, fibril toxicity and fibril spreading.
Collapse
|
30
|
Kazakova O, Giniyatullina G, Babkov D, Wimmer Z. From Marine Metabolites to the Drugs of the Future: Squalamine, Trodusquemine, Their Steroid and Triterpene Analogues. Int J Mol Sci 2022; 23:ijms23031075. [PMID: 35162998 PMCID: PMC8834734 DOI: 10.3390/ijms23031075] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
This review comprehensively describes the recent advances in the synthesis and pharmacological evaluation of steroid polyamines squalamine, trodusquemine, ceragenins, claramine, and their diverse analogs and derivatives, with a special focus on their complete synthesis from cholic acids, as well as an antibacterial and antiviral, neuroprotective, antiangiogenic, antitumor, antiobesity and weight-loss activity, antiatherogenic, regenerative, and anxiolytic properties. Trodusquemine is the most-studied small-molecule allosteric PTP1B inhibitor. The discovery of squalamine as the first representative of a previously unknown class of natural antibiotics of animal origin stimulated extensive research of terpenoids (especially triterpenoids) comprising polyamine fragments. During the last decade, this new class of biologically active semisynthetic natural product derivatives demonstrated the possibility to form supramolecular networks, which opens up many possibilities for the use of such structures for drug delivery systems in serum or other body fluids.
Collapse
Affiliation(s)
- Oxana Kazakova
- Ufa Institute of Chemistry, UFA Federal Research Centre of the Russian Academy of Sciences, Pr. Oktyabrya, 450054 Ufa, Russia;
- Correspondence:
| | - Gulnara Giniyatullina
- Ufa Institute of Chemistry, UFA Federal Research Centre of the Russian Academy of Sciences, Pr. Oktyabrya, 450054 Ufa, Russia;
| | - Denis Babkov
- Laboratory of Metabotropic Drugs, Scientific Center for Innovative Drugs, Volgograd State Medical University, Novorossiyskaya St. 39, 400087 Volgograd, Russia;
| | - Zdenek Wimmer
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology in Prague, Technicka’ 5, Prague 6, 16628 Prague, Czech Republic;
| |
Collapse
|
31
|
Nguyen PH, Tufféry P, Derreumaux P. Dynamics of Amyloid Formation from Simplified Representation to Atomistic Simulations. Methods Mol Biol 2022; 2405:95-113. [PMID: 35298810 DOI: 10.1007/978-1-0716-1855-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Amyloid fibril formation is an intrinsic property of short peptides, non-disease proteins, and proteins associated with neurodegenerative diseases. Aggregates of the Aβ and tau proteins, the α-synuclein protein, and the prion protein are observed in the brain of Alzheimer's, Parkinson's, and prion disease patients, respectively. Due to the transient short-range and long-range interactions of all species and their high aggregation propensities, the conformational ensemble of these devastating proteins, the exception being for the monomeric prion protein, remains elusive by standard structural biology methods in bulk solution and in lipid membranes. To overcome these limitations, an increasing number of simulations using different sampling methods and protein models have been performed. In this chapter, we first review our main contributions to the field of amyloid protein simulations aimed at understanding the early aggregation steps of short linear amyloid peptides, the conformational ensemble of the Aβ40/42 dimers in bulk solution, and the stability of Aβ aggregates in lipid membrane models. Then we focus on our studies on the interactions of amyloid peptides/inhibitors to prevent aggregation, and long amyloid sequences, including new results on a monomeric tau construct.
Collapse
Affiliation(s)
- Phuong Hoang Nguyen
- Laboratoire de Biochimie Théorique, CNRS, Université de Paris, UPR 9080, Paris, France
- Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, PSL Research University, Paris, France
| | - Pierre Tufféry
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, RPBS, Paris, France
| | - Philippe Derreumaux
- Laboratoire de Biochimie Théorique, CNRS, Université de Paris, UPR 9080, Paris, France.
- Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, PSL Research University, Paris, France.
| |
Collapse
|
32
|
Limbocker R, Errico S, Barbut D, Knowles TPJ, Vendruscolo M, Chiti F, Zasloff M. Squalamine and trodusquemine: two natural products for neurodegenerative diseases, from physical chemistry to the clinic. Nat Prod Rep 2021; 39:742-753. [PMID: 34698757 DOI: 10.1039/d1np00042j] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Covering: 1993 to 2021 (mainly 2017-2021)Alzheimer's and Parkinson's diseases are neurodegenerative conditions affecting over 50 million people worldwide. Since these disorders are still largely intractable pharmacologically, discovering effective treatments is of great urgency and importance. These conditions are characteristically associated with the aberrant deposition of proteinaceous aggregates in the brain, and with the formation of metastable intermediates known as protein misfolded oligomers that play a central role in their aetiology. In this Highlight article, we review the evidence at the physicochemical, cellular, animal model and clinical levels on how the natural products squalamine and trodusquemine offer promising opportunities for chronic treatments for these progressive conditions by preventing both the formation of neurotoxic oligomers and their interaction with cell membranes.
Collapse
Affiliation(s)
- Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Silvia Errico
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy. .,Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK.
| | - Denise Barbut
- Enterin Inc., 3624 Market Street, Philadelphia, Pennsylvania 19104, USA
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK. .,Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK.
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy.
| | - Michael Zasloff
- Enterin Inc., 3624 Market Street, Philadelphia, Pennsylvania 19104, USA.,MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, DC 20010, USA.
| |
Collapse
|
33
|
Unzipping the Secrets of Amyloid Disassembly by the Human Disaggregase. Cells 2021; 10:cells10102745. [PMID: 34685723 PMCID: PMC8534776 DOI: 10.3390/cells10102745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases (NDs) are increasingly positioned as leading causes of global deaths. The accelerated aging of the population and its strong relationship with neurodegeneration forecast these pathologies as a huge global health problem in the upcoming years. In this scenario, there is an urgent need for understanding the basic molecular mechanisms associated with such diseases. A major molecular hallmark of most NDs is the accumulation of insoluble and toxic protein aggregates, known as amyloids, in extracellular or intracellular deposits. Here, we review the current knowledge on how molecular chaperones, and more specifically a ternary protein complex referred to as the human disaggregase, deals with amyloids. This machinery, composed of the constitutive Hsp70 (Hsc70), the class B J-protein DnaJB1 and the nucleotide exchange factor Apg2 (Hsp110), disassembles amyloids of α-synuclein implicated in Parkinson’s disease as well as of other disease-associated proteins such as tau and huntingtin. We highlight recent studies that have led to the dissection of the mechanism used by this chaperone system to perform its disaggregase activity. We also discuss whether this chaperone-mediated disassembly mechanism could be used to solubilize other amyloidogenic substrates. Finally, we evaluate the implications of the chaperone system in amyloid clearance and associated toxicity, which could be critical for the development of new therapies.
Collapse
|
34
|
Errico S, Ramshini H, Capitini C, Canale C, Spaziano M, Barbut D, Calamai M, Zasloff M, Oropesa-Nuñez R, Vendruscolo M, Chiti F. Quantitative Measurement of the Affinity of Toxic and Nontoxic Misfolded Protein Oligomers for Lipid Bilayers and of its Modulation by Lipid Composition and Trodusquemine. ACS Chem Neurosci 2021; 12:3189-3202. [PMID: 34382791 PMCID: PMC8414483 DOI: 10.1021/acschemneuro.1c00327] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Many neurodegenerative diseases are associated with the self-assembly of peptides and proteins into fibrillar aggregates. Soluble misfolded oligomers formed during the aggregation process, or released by mature fibrils, play a relevant role in neurodegenerative processes through their interactions with neuronal membranes. However, the determinants of the cytotoxicity of these oligomers are still unclear. Here we used liposomes and toxic and nontoxic oligomers formed by the same protein to measure quantitatively the affinity of the two oligomeric species for lipid membranes. To this aim, we quantified the perturbation to the lipid membranes caused by the two oligomers by using the fluorescence quenching of two probes embedded in the polar and apolar regions of the lipid membranes and a well-defined protein-oligomer binding assay using fluorescently labeled oligomers to determine the Stern-Volmer and dissociation constants, respectively. With both approaches, we found that the toxic oligomers have a membrane affinity 20-25 times higher than that of nontoxic oligomers. Circular dichroism, intrinsic fluorescence, and FRET indicated that neither oligomer type changes its structure upon membrane interaction. Using liposomes enriched with trodusquemine, a potential small molecule drug known to penetrate lipid membranes and make them refractory to toxic oligomers, we found that the membrane affinity of the oligomers was remarkably lower. At protective concentrations of the small molecule, the binding of the oligomers to the lipid membranes was fully prevented. Furthermore, the affinity of the toxic oligomers for the lipid membranes was found to increase and slightly decrease with GM1 ganglioside and cholesterol content, respectively, indicating that physicochemical properties of lipid membranes modulate their affinity for misfolded oligomeric species.
Collapse
Affiliation(s)
- Silvia Errico
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Hassan Ramshini
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
- Department
of Biology, Payame Noor University, Tehran 19395-4697, Islamic Republic of Iran
| | - Claudia Capitini
- European
Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino 50019, Italy
- Department
of Physics and Astronomy, University of
Florence, Sesto
Fiorentino 50019, Italy
| | - Claudio Canale
- Department
of Physics, University of Genoa, Genoa 16146, Italy
| | - Martina Spaziano
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Denise Barbut
- Enterin
Inc., 2005 Market Street, Philadelphia, Pennsylvania 19103, United States
| | - Martino Calamai
- European
Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino 50019, Italy
- National
Institute of Optics, National Research Council
of Italy (CNR), Florence 50125, Italy
| | - Michael Zasloff
- Enterin
Inc., 2005 Market Street, Philadelphia, Pennsylvania 19103, United States
- MedStar-Georgetown
Transplant Institute, Georgetown University
School of Medicine, Washington D.C. 20007, United States
| | - Reinier Oropesa-Nuñez
- Department
of Materials Science and Engineering, Uppsala
University, Uppsala SE-751 03, Sweden
| | - Michele Vendruscolo
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Fabrizio Chiti
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| |
Collapse
|
35
|
Yahi N, Di Scala C, Chahinian H, Fantini J. Innovative treatment targeting gangliosides aimed at blocking the formation of neurotoxic α-synuclein oligomers in Parkinson's disease. Glycoconj J 2021; 39:1-11. [PMID: 34328594 DOI: 10.1007/s10719-021-10012-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a major neurodegenerative disorder which exhibits many of the characteristics of a pandemic. Current therapeutic strategies are centered on the dopaminergic system, with limited efficacy, so that a treatment that has a direct impact on the underlying disease pathogenesis is urgently needed. Although α-synuclein is a privileged target for such therapies, this protein has been in the past wrongly considered as exclusively intracellular, so that the impact of paracrine neurotoxicity mechanisms in PD have been largely ignored. In this article we review the data showing that lipid rafts act as plasma membrane machineries for the formation of α-synuclein pore-like oligomers which trigger an increase of intracellular Ca2+. This Ca2+ influx is responsible for a self-sustained cascade of neurotoxic events, including mitochondrial oxidative stress, tau phosphorylation, Ca2+ release from the endoplasmic reticulum, Lewy body formation, and extracellular release of α-synuclein in exosomes. The first step of this cascade is the binding of α-synuclein to lipid raft gangliosides, suggesting that PD should be considered as both a proteinopathy and a ganglioside membrane disorder lipidopathy. Accordingly, blocking α-synuclein-ganglioside interactions should annihilate the whole neurotoxic cascade and stop disease progression. A pipeline of anti-oligomer molecules is under development, among which an in-silico designed synthetic peptide AmyP53 which is the first drug targeting gangliosides and thus able to prevent the formation of α-synuclein oligomers and all downstream neurotoxicity. These new therapeutic avenues challenge the current symptomatic approaches by finally targeting the root cause of PD through a long-awaited paradigm shift.
Collapse
Affiliation(s)
- Nouara Yahi
- INSERM UMR_S 1072, Aix-Marseille Université, 13015, Marseille, France
| | - Coralie Di Scala
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Henri Chahinian
- INSERM UMR_S 1072, Aix-Marseille Université, 13015, Marseille, France
| | - Jacques Fantini
- INSERM UMR_S 1072, Aix-Marseille Université, 13015, Marseille, France.
| |
Collapse
|
36
|
Perni M, Mannini B, Xu CK, Kumita JR, Dobson CM, Chiti F, Vendruscolo M. Exogenous misfolded protein oligomers can cross the intestinal barrier and cause a disease phenotype in C. elegans. Sci Rep 2021; 11:14391. [PMID: 34257326 PMCID: PMC8277765 DOI: 10.1038/s41598-021-93527-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Misfolded protein oligomers are increasingly recognized as highly cytotoxic agents in a wide range of human disorders associated with protein aggregation. In this study, we assessed the possible uptake and resulting toxic effects of model protein oligomers administered to C. elegans through the culture medium. We used an automated machine-vision, high-throughput screening procedure to monitor the phenotypic changes in the worms, in combination with confocal microscopy to monitor the diffusion of the oligomers, and oxidative stress assays to detect their toxic effects. Our results suggest that the oligomers can diffuse from the intestinal lumen to other tissues, resulting in a disease phenotype. We also observed that pre-incubation of the oligomers with a molecular chaperone (αB-crystallin) or a small molecule inhibitor of protein aggregation (squalamine), reduced the oligomer absorption. These results indicate that exogenous misfolded protein oligomers can be taken up by the worms from their environment and spread across tissues, giving rise to pathological effects in regions distant from their place of absorbance.
Collapse
Affiliation(s)
- Michele Perni
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Benedetta Mannini
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Catherine K Xu
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Janet R Kumita
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
37
|
Limbocker R, Staats R, Chia S, Ruggeri FS, Mannini B, Xu CK, Perni M, Cascella R, Bigi A, Sasser LR, Block NR, Wright AK, Kreiser RP, Custy ET, Meisl G, Errico S, Habchi J, Flagmeier P, Kartanas T, Hollows JE, Nguyen LT, LeForte K, Barbut D, Kumita JR, Cecchi C, Zasloff M, Knowles TPJ, Dobson CM, Chiti F, Vendruscolo M. Squalamine and Its Derivatives Modulate the Aggregation of Amyloid-β and α-Synuclein and Suppress the Toxicity of Their Oligomers. Front Neurosci 2021; 15:680026. [PMID: 34220435 PMCID: PMC8249941 DOI: 10.3389/fnins.2021.680026] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
The aberrant aggregation of proteins is a key molecular event in the development and progression of a wide range of neurodegenerative disorders. We have shown previously that squalamine and trodusquemine, two natural products in the aminosterol class, can modulate the aggregation of the amyloid-β peptide (Aβ) and of α-synuclein (αS), which are associated with Alzheimer's and Parkinson's diseases. In this work, we expand our previous analyses to two squalamine derivatives, des-squalamine and α-squalamine, obtaining further insights into the mechanism by which aminosterols modulate Aβ and αS aggregation. We then characterize the ability of these small molecules to alter the physicochemical properties of stabilized oligomeric species in vitro and to suppress the toxicity of these aggregates to varying degrees toward human neuroblastoma cells. We found that, despite the fact that these aminosterols exert opposing effects on Aβ and αS aggregation under the conditions that we tested, the modifications that they induced to the toxicity of oligomers were similar. Our results indicate that the suppression of toxicity is mediated by the displacement of toxic oligomeric species from cellular membranes by the aminosterols. This study, thus, provides evidence that aminosterols could be rationally optimized in drug discovery programs to target oligomer toxicity in Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Ryan Limbocker
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Roxine Staats
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Sean Chia
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Francesco S. Ruggeri
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Laboratory of Organic Chemistry, Wageningen University, Wageningen, Netherlands
- Laboratory of Physical Chemistry, Wageningen University, Wageningen, Netherlands
| | - Benedetta Mannini
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Catherine K. Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Michele Perni
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Liam R. Sasser
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Natalie R. Block
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Aidan K. Wright
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Ryan P. Kreiser
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Edward T. Custy
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Silvia Errico
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Johnny Habchi
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Patrick Flagmeier
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Tadas Kartanas
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Jared E. Hollows
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Lam T. Nguyen
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | - Kathleen LeForte
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, United States
| | | | - Janet R. Kumita
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Michael Zasloff
- Enterin Inc., Philadelphia, PA, United States
- MedStar Georgetown Transplant Institute, School of Medicine, Georgetown University, Washington, DC, United States
| | - Tuomas P. J. Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - Christopher M. Dobson
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
38
|
Zhang L, Qin Z, Sharmin F, Lin W, Ricke KM, Zasloff MA, Stewart AFR, Chen HH. Tyrosine phosphatase PTP1B impairs presynaptic NMDA receptor-mediated plasticity in a mouse model of Alzheimer's disease. Neurobiol Dis 2021; 156:105402. [PMID: 34044147 DOI: 10.1016/j.nbd.2021.105402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Mutations in the beta-amyloid protein (APP) cause familial Alzheimer's disease. In hAPP-J20 mice expressing mutant APP, pharmacological inhibition or genetic ablation of the tyrosine phosphatase PTP1B prevents CA3 hippocampus neuron loss and cognitive decline. However, how targeting PTP1B affects the cellular mechanisms underlying these cognitive deficits remains unknown. Changes in synaptic strength at the hippocampus can affect information processing for learning and memory. While prior studies have focused on post-synaptic mechanisms to account for synaptic deficits in Alzheimer's disease models, presynaptic mechanisms may also be affected. Here, using whole cell patch-clamp recording, coefficient of variation (CV) analysis suggested a profound presynaptic deficit in long-term potentiation (LTP) of CA3:CA1 synapses in hAPP-J20 mice. While the membrane-impermeable ionotropic NMDA receptor (NMDAR) blocker norketamine in the post-synaptic recording electrode had no effect on LTP, additional bath application of the ionotropic NMDAR blockers MK801 could replicate the deficit in LTP in wild type mice. In contrast to LTP, the paired-pulse ratio and short-term facilitation (STF) were aberrantly increased in hAPP-J20 mice. These synaptic deficits in hAPP-J20 mice were associated with reduced phosphorylation of NMDAR GluN2B and the synaptic vesicle recycling protein NSF (N-ethylmaleimide sensitive factor). Phosphorylation of both proteins, together with synaptic plasticity and cognitive function, were restored by PTP1B ablation or inhibition by the PTP1B-selective inhibitor Trodusquemine. Taken together, our results indicate that PTP1B impairs presynaptic NMDAR-mediated synaptic plasticity required for spatial learning in a mouse model of Alzheimer's disease. Since Trodusquemine has undergone phase 1/2 clinical trials to treat obesity, it could be repurposed to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Li Zhang
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada
| | - Zhaohong Qin
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada
| | - Fariba Sharmin
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Wei Lin
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada
| | - Konrad M Ricke
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada
| | - Michael A Zasloff
- Georgetown University School of Medicine, MedStar Georgetown Transplant Institute, Washington, DC, 2007, USA
| | - Alexandre F R Stewart
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada.
| | - Hsiao-Huei Chen
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
39
|
Cascella R, Cecchi C. Calcium Dyshomeostasis in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22094914. [PMID: 34066371 PMCID: PMC8124842 DOI: 10.3390/ijms22094914] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 01/12/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disorder that is characterized by amyloid β-protein deposition in senile plaques, neurofibrillary tangles consisting of abnormally phosphorylated tau protein, and neuronal loss leading to cognitive decline and dementia. Despite extensive research, the exact mechanisms underlying AD remain unknown and effective treatment is not available. Many hypotheses have been proposed to explain AD pathophysiology; however, there is general consensus that the abnormal aggregation of the amyloid β peptide (Aβ) is the initial event triggering a pathogenic cascade of degenerating events in cholinergic neurons. The dysregulation of calcium homeostasis has been studied considerably to clarify the mechanisms of neurodegeneration induced by Aβ. Intracellular calcium acts as a second messenger and plays a key role in the regulation of neuronal functions, such as neural growth and differentiation, action potential, and synaptic plasticity. The calcium hypothesis of AD posits that activation of the amyloidogenic pathway affects neuronal Ca2+ homeostasis and the mechanisms responsible for learning and memory. Aβ can disrupt Ca2+ signaling through several mechanisms, by increasing the influx of Ca2+ from the extracellular space and by activating its release from intracellular stores. Here, we review the different molecular mechanisms and receptors involved in calcium dysregulation in AD and possible therapeutic strategies for improving the treatment.
Collapse
|
40
|
Khoury ZH, Salameh F. Trodusquemine: Potential Utility in Wound Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021. [DOI: 10.1007/s40883-021-00211-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
41
|
Perni M, van der Goot A, Limbocker R, van Ham TJ, Aprile FA, Xu CK, Flagmeier P, Thijssen K, Sormanni P, Fusco G, Chen SW, Challa PK, Kirkegaard JB, Laine RF, Ma KY, Müller MBD, Sinnige T, Kumita JR, Cohen SIA, Seinstra R, Kaminski Schierle GS, Kaminski CF, Barbut D, De Simone A, Knowles TPJ, Zasloff M, Nollen EAA, Vendruscolo M, Dobson CM. Comparative Studies in the A30P and A53T α-Synuclein C. elegans Strains to Investigate the Molecular Origins of Parkinson's Disease. Front Cell Dev Biol 2021; 9:552549. [PMID: 33829010 PMCID: PMC8019828 DOI: 10.3389/fcell.2021.552549] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 02/16/2021] [Indexed: 02/02/2023] Open
Abstract
The aggregation of α-synuclein is a hallmark of Parkinson's disease (PD) and a variety of related neurological disorders. A number of mutations in this protein, including A30P and A53T, are associated with familial forms of the disease. Patients carrying the A30P mutation typically exhibit a similar age of onset and symptoms as sporadic PD, while those carrying the A53T mutation generally have an earlier age of onset and an accelerated progression. We report two C. elegans models of PD (PDA30P and PDA53T), which express these mutational variants in the muscle cells, and probed their behavior relative to animals expressing the wild-type protein (PDWT). PDA30P worms showed a reduced speed of movement and an increased paralysis rate, control worms, but no change in the frequency of body bends. By contrast, in PDA53T worms both speed and frequency of body bends were significantly decreased, and paralysis rate was increased. α-Synuclein was also observed to be less well localized into aggregates in PDA30P worms compared to PDA53T and PDWT worms, and amyloid-like features were evident later in the life of the animals, despite comparable levels of expression of α-synuclein. Furthermore, squalamine, a natural product currently in clinical trials for treating symptomatic aspects of PD, was found to reduce significantly the aggregation of α-synuclein and its associated toxicity in PDA53T and PDWT worms, but had less marked effects in PDA30P. In addition, using an antibody that targets the N-terminal region of α-synuclein, we observed a suppression of toxicity in PDA30P, PDA53T and PDWT worms. These results illustrate the use of these two C. elegans models in fundamental and applied PD research.
Collapse
Affiliation(s)
- Michele Perni
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Annemieke van der Goot
- University Medical Centre Groningen, European Research Institute for the Biology of Aging, University of Groningen, Groningen, Netherlands
| | - Ryan Limbocker
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom,Department of Chemistry and Life Science, United States Military Academy, West Point, NY, United States
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Francesco A. Aprile
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Catherine K. Xu
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Patrick Flagmeier
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Karen Thijssen
- University Medical Centre Groningen, European Research Institute for the Biology of Aging, University of Groningen, Groningen, Netherlands
| | - Pietro Sormanni
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Giuliana Fusco
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Serene W. Chen
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Pavan K. Challa
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Julius B. Kirkegaard
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
| | - Romain F. Laine
- MRC Laboratory for Molecular Cell Biology (LMCB) University College London, London, United Kingdom
| | - Kai Yu Ma
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom,University Medical Centre Groningen, European Research Institute for the Biology of Aging, University of Groningen, Groningen, Netherlands
| | - Martin B. D. Müller
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom,University Medical Centre Groningen, European Research Institute for the Biology of Aging, University of Groningen, Groningen, Netherlands
| | - Tessa Sinnige
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Janet R. Kumita
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Samuel I. A. Cohen
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Renée Seinstra
- University Medical Centre Groningen, European Research Institute for the Biology of Aging, University of Groningen, Groningen, Netherlands
| | | | - Clemens F. Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Denise Barbut
- MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, DC, United States
| | - Alfonso De Simone
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Tuomas P. J. Knowles
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Michael Zasloff
- MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, DC, United States
| | - Ellen A. A. Nollen
- University Medical Centre Groningen, European Research Institute for the Biology of Aging, University of Groningen, Groningen, Netherlands,*Correspondence: Ellen A. A. Nollen
| | - Michele Vendruscolo
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom,Michele Vendruscolo
| | - Christopher M. Dobson
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
42
|
Cascella R, Chen SW, Bigi A, Camino JD, Xu CK, Dobson CM, Chiti F, Cremades N, Cecchi C. The release of toxic oligomers from α-synuclein fibrils induces dysfunction in neuronal cells. Nat Commun 2021; 12:1814. [PMID: 33753734 PMCID: PMC7985515 DOI: 10.1038/s41467-021-21937-3] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
The self-assembly of α-synuclein (αS) into intraneuronal inclusion bodies is a key characteristic of Parkinson's disease. To define the nature of the species giving rise to neuronal damage, we have investigated the mechanism of action of the main αS populations that have been observed to form progressively during fibril growth. The αS fibrils release soluble prefibrillar oligomeric species with cross-β structure and solvent-exposed hydrophobic clusters. αS prefibrillar oligomers are efficient in crossing and permeabilize neuronal membranes, causing cellular insults. Short fibrils are more neurotoxic than long fibrils due to the higher proportion of fibrillar ends, resulting in a rapid release of oligomers. The kinetics of released αS oligomers match the observed kinetics of toxicity in cellular systems. In addition to previous evidence that αS fibrils can spread in different brain areas, our in vitro results reveal that αS fibrils can also release oligomeric species responsible for an immediate dysfunction of the neurons in the vicinity of these species.
Collapse
Affiliation(s)
- Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Serene W Chen
- Department of Life Science, Imperial College London, London, UK
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - José D Camino
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Joint Unit BIFI-Institute of Physical Chemistry "Rocasolano" (CSIC), University of Zaragoza, Zaragoza, Spain
| | - Catherine K Xu
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Nunilo Cremades
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Joint Unit BIFI-Institute of Physical Chemistry "Rocasolano" (CSIC), University of Zaragoza, Zaragoza, Spain.
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy.
| |
Collapse
|
43
|
Fani G, Mannini B, Vecchi G, Cascella R, Cecchi C, Dobson CM, Vendruscolo M, Chiti F. Aβ Oligomers Dysregulate Calcium Homeostasis by Mechanosensitive Activation of AMPA and NMDA Receptors. ACS Chem Neurosci 2021; 12:766-781. [PMID: 33538575 PMCID: PMC7898266 DOI: 10.1021/acschemneuro.0c00811] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease, which is the most common form of dementia, is characterized by the aggregation of the amyloid β peptide (Aβ) and by an impairment of calcium homeostasis caused by excessive activation of glutamatergic receptors (excitotoxicity). Here, we studied the effects on calcium homeostasis caused by the formation of Aβ oligomeric assemblies. We found that Aβ oligomers cause a rapid influx of calcium ions (Ca2+) across the cell membrane by rapidly activating extrasynaptic N-methyl-d-aspartate (NMDA) receptors and, to a lower extent, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. We also observed, however, that misfolded oligomers do not interact directly with these receptors. Further experiments with lysophosphatidylcholine and arachidonic acid, which cause membrane compression and stretch, respectively, indicated that these receptors are activated through a change in membrane tension induced by the oligomers and transmitted mechanically to the receptors via the lipid bilayer. Indeed, lysophosphatidylcholine is able to neutralize the oligomer-induced activation of the NMDA receptors, whereas arachidonic acid activates the receptors similarly to the oligomers with no additive effects. An increased rotational freedom observed for a fluorescent probe embedded within the membrane in the presence of the oligomers also indicates a membrane stretch. These results reveal a mechanism of toxicity of Aβ oligomers in Alzheimer's disease through the perturbation of the mechanical properties of lipid membranes sensed by NMDA and AMPA receptors.
Collapse
Affiliation(s)
- Giulia Fani
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
- Centre
for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Benedetta Mannini
- Centre
for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Giulia Vecchi
- Centre
for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Roberta Cascella
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Cristina Cecchi
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Christopher M. Dobson
- Centre
for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Michele Vendruscolo
- Centre
for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Fabrizio Chiti
- Department
of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
44
|
Cataldi R, Chia S, Pisani K, Ruggeri FS, Xu CK, Šneideris T, Perni M, Sarwat S, Joshi P, Kumita JR, Linse S, Habchi J, Knowles TPJ, Mannini B, Dobson CM, Vendruscolo M. A dopamine metabolite stabilizes neurotoxic amyloid-β oligomers. Commun Biol 2021; 4:19. [PMID: 33398040 PMCID: PMC7782527 DOI: 10.1038/s42003-020-01490-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022] Open
Abstract
Aberrant soluble oligomers formed by the amyloid-β peptide (Aβ) are major pathogenic agents in the onset and progression of Alzheimer's disease. A variety of biomolecules can influence the formation of these oligomers in the brain, although their mechanisms of action are still largely unknown. Here, we studied the effects on Aβ aggregation of DOPAL, a reactive catecholaldehyde intermediate of dopamine metabolism. We found that DOPAL is able to stabilize Aβ oligomeric species, including dimers and trimers, that exert toxic effects on human neuroblastoma cells, in particular increasing cytosolic calcium levels and promoting the generation of reactive oxygen species. These results reveal an interplay between Aβ aggregation and key biochemical processes regulating cellular homeostasis in the brain.
Collapse
Affiliation(s)
- Rodrigo Cataldi
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Sean Chia
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Katarina Pisani
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Francesco S Ruggeri
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Catherine K Xu
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Tomas Šneideris
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, 10257, Lithuania
| | - Michele Perni
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Sunehera Sarwat
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Priyanka Joshi
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Janet R Kumita
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Sara Linse
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Lund, Sweden
| | - Johnny Habchi
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
| | - Benedetta Mannini
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
45
|
Musteikytė G, Jayaram AK, Xu CK, Vendruscolo M, Krainer G, Knowles TPJ. Interactions of α-synuclein oligomers with lipid membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183536. [PMID: 33373595 DOI: 10.1016/j.bbamem.2020.183536] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/24/2022]
Abstract
Parkinson's disease is an increasingly prevalent and currently incurable neurodegenerative disorder. At the molecular level, this disease is characterized by the formation of aberrant intracellular protein deposits known as Lewy bodies. Oligomeric forms of the protein α-synuclein (αS), which are believed to be both intermediates and by-products of Lewy body formation, are considered to be the main pathogenic species. Interactions of such oligomers with lipid membranes are increasingly emerging as a major molecular pathway underpinning their toxicity. Here we review recent progress in our understanding of the interactions of αS oligomers with lipid membranes. We highlight key structural and biophysical features of αS oligomers, the effects of these features on αS oligomer membrane binding properties, and resultant implications for understanding the etiology of Parkinson's disease. We discuss mechanistic modes of αS oligomer-lipid membrane interactions and the effects of environmental factors to such modes. Finally, we provide an overview of the current understanding of the main molecular determinants of αS oligomer toxicity in vivo.
Collapse
Affiliation(s)
- Greta Musteikytė
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Akhila K Jayaram
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom; Cavendish Laboratory, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Catherine K Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Georg Krainer
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom; Cavendish Laboratory, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom.
| |
Collapse
|
46
|
Errico S, Lucchesi G, Odino D, Muscat S, Capitini C, Bugelli C, Canale C, Ferrando R, Grasso G, Barbut D, Calamai M, Danani A, Zasloff M, Relini A, Caminati G, Vendruscolo M, Chiti F. Making biological membrane resistant to the toxicity of misfolded protein oligomers: a lesson from trodusquemine. NANOSCALE 2020; 12:22596-22614. [PMID: 33150350 DOI: 10.1039/d0nr05285j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Trodusquemine is an aminosterol known to prevent the binding of misfolded protein oligomers to cell membranes and to reduce their toxicity in a wide range of neurodegenerative diseases. Its precise mechanism of action, however, remains unclear. To investigate this mechanism, we performed confocal microscopy, fluorescence resonance energy transfer (FRET) and nuclear magnetic resonance (NMR) measurements, which revealed a strong binding of trodusquemine to large unilamellar vesicles (LUVs) and neuroblastoma cell membranes. Then, by combining quartz crystal microbalance (QCM), fluorescence quenching and anisotropy, and molecular dynamics (MD) simulations, we found that trodusquemine localises within, and penetrates, the polar region of lipid bilayer. This binding behaviour causes a decrease of the negative charge of the bilayer, as observed through ζ potential measurements, an increment in the mechanical resistance of the bilayer, as revealed by measurements of the breakthrough force applied with AFM and ζ potential measurements at high temperature, and a rearrangement of the spatial distances between ganglioside and cholesterol molecules in the LUVs, as determined by FRET measurements. These physicochemical changes are all known to impair the interaction of misfolded oligomers with cell membranes, protecting them from their toxicity. Taken together, our results illustrate how the incorporation in cell membranes of sterol molecules modified by the addition of polyamine tails leads to the modulation of physicochemical properties of the cell membranes themselves, making them more resistant to protein aggregates associated with neurodegeneration. More generally, they suggest that therapeutic strategies can be developed to reinforce cell membranes against protein misfolded assemblies.
Collapse
Affiliation(s)
- Silvia Errico
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kreiser RP, Wright AK, Block NR, Hollows JE, Nguyen LT, LeForte K, Mannini B, Vendruscolo M, Limbocker R. Therapeutic Strategies to Reduce the Toxicity of Misfolded Protein Oligomers. Int J Mol Sci 2020; 21:ijms21228651. [PMID: 33212787 PMCID: PMC7696907 DOI: 10.3390/ijms21228651] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
The aberrant aggregation of proteins is implicated in the onset and pathogenesis of a wide range of neurodegenerative disorders, including Alzheimer’s and Parkinson’s diseases. Mounting evidence indicates that misfolded protein oligomers produced as intermediates in the aggregation process are potent neurotoxic agents in these diseases. Because of the transient and heterogeneous nature of these elusive aggregates, however, it has proven challenging to develop therapeutics that can effectively target them. Here, we review approaches aimed at reducing oligomer toxicity, including (1) modulating the oligomer populations (e.g., by altering the kinetics of aggregation by inhibiting, enhancing, or redirecting the process), (2) modulating the oligomer properties (e.g., through the size–hydrophobicity–toxicity relationship), (3) modulating the oligomer interactions (e.g., by protecting cell membranes by displacing oligomers), and (4) reducing oligomer toxicity by potentiating the protein homeostasis system. We analyze examples of these complementary approaches, which may lead to the development of compounds capable of preventing or treating neurodegenerative disorders associated with protein aggregation.
Collapse
Affiliation(s)
- Ryan P. Kreiser
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA; (R.P.K.); (A.K.W.); (N.R.B.); (J.E.H.); (L.T.N.); (K.L.)
| | - Aidan K. Wright
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA; (R.P.K.); (A.K.W.); (N.R.B.); (J.E.H.); (L.T.N.); (K.L.)
| | - Natalie R. Block
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA; (R.P.K.); (A.K.W.); (N.R.B.); (J.E.H.); (L.T.N.); (K.L.)
| | - Jared E. Hollows
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA; (R.P.K.); (A.K.W.); (N.R.B.); (J.E.H.); (L.T.N.); (K.L.)
| | - Lam T. Nguyen
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA; (R.P.K.); (A.K.W.); (N.R.B.); (J.E.H.); (L.T.N.); (K.L.)
| | - Kathleen LeForte
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA; (R.P.K.); (A.K.W.); (N.R.B.); (J.E.H.); (L.T.N.); (K.L.)
| | - Benedetta Mannini
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK;
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK;
- Correspondence: (M.V.); (R.L.)
| | - Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA; (R.P.K.); (A.K.W.); (N.R.B.); (J.E.H.); (L.T.N.); (K.L.)
- Correspondence: (M.V.); (R.L.)
| |
Collapse
|
48
|
Du XY, Xie XX, Liu RT. The Role of α-Synuclein Oligomers in Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21228645. [PMID: 33212758 PMCID: PMC7697105 DOI: 10.3390/ijms21228645] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
α-synuclein (α-syn) is a protein associated with the pathogenesis of Parkinson’s disease (PD), the second most common neurodegeneration disease with no effective treatment. However, how α-syn drives the pathology of PD remains elusive. Recent studies suggest that α-syn oligomers are the primary cause of neurotoxicity and play a critical role in PD. In this review, we discuss the process of α-syn oligomers formation and the current understanding of the structures of oligomers. We also describe seed and propagation effects of oligomeric forms of α-syn. Then, we summarize the mechanism by which α-syn oligomers exert neurotoxicity and promote neurodegeneration, including mitochondrial dysfunction, endoplasmic reticulum stress, proteostasis dysregulation, synaptic impairment, cell apoptosis and neuroinflammation. Finally, we investigate treatment regimens targeting α-syn oligomers at present. Further research is needed to understand the structure and toxicity mechanism of different types of oligomers, so as to provide theoretical basis for the treatment of PD.
Collapse
Affiliation(s)
- Xiao-yu Du
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing 100190, China; (X.-y.D.); (X.-x.X.)
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi-xiu Xie
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing 100190, China; (X.-y.D.); (X.-x.X.)
| | - Rui-tian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing 100190, China; (X.-y.D.); (X.-x.X.)
- Correspondence: ; Tel.: +86-10-82545017
| |
Collapse
|