1
|
Kung YH, Chang CY, Lai YR, Li JX, How SC. Examining the inhibitory potency of metal polyphenolic network-coated silver nanoparticles against amyloid fibrillogenesis of lysozyme. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 327:125375. [PMID: 39527883 DOI: 10.1016/j.saa.2024.125375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
There are currently over forty degenerative diseases that are correlated with abnormal accumulation of peptide/protein aggregates in the human body, such as Alzheimer's disease. Due to their unique physiochemical properties (e.g., small size, large surface-to-volume ratio, and facile surface modification), silver nanoparticles (AgNPs) have been considered potential substrates for designing inhibitors against amyloid fibrillogenesis. Metal polyphenolic network (MPN) that combines the characteristics of organic and inorganic components has been used to suppress amyloid fibril formation. This study is aimed at investigating the effects of MPN-coated AgNPs (MPN-AgNPs) on the in vitro amyloid fibrillogenesis of hen lysozyme (HEWL). The two types of MPN-AgNPs (Zn/MPN-AgNPs and Co/MPN-AgNPs) were synthesized through chemical reduction and metal chelation, and their particle sizes were determined to be in the range of 40-60 nm. The characterization of MPN-AgNPs by ζ-potential and transmission electron microscopy showed that the MPN-AgNPs had negative surface charge and spherical-shaped morphology. Furthermore, the elemental analysis demonstrated that the MPN was uniformly coated on the surface of AgNPs. The thioflavin T fluorescence results revealed that the Co/MPN-AgNPs showed a better percent of inhibition compared to Zn/MPN-AgNPs and TA-AgNPs. The kinetics data of amyloid fibril formation in the presence of MPN-AgNPs were analyzed using the sigmoidal curve, showing that the MPN-AgNPs reduced fibril growth rate and prolonged lag time. Our findings also demonstrated that MPN-AgNPs could effectively suppress HEWL aggregation upon binding to aggregation-prone regions. The quenching of intrinsic fluorescence of HEWL by MPN-AgNPs was found to be a static type. Moreover, the fluorescence quenching data were analyzed using the modified Stern-Volmer equation to determine the number of binding sites. Notably, our findings indicated that the binding between HEWL and MPN-AgNPs was mainly governed by hydrophobic interaction. This work offers an excellent example of utilizing MPN-based materials as anti-aggregating/anti-fibrillogenic nano-drugs for the treatment of amyloidosis.
Collapse
Affiliation(s)
- Yu-Hsuan Kung
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Chia-Yu Chang
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - You-Ren Lai
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Jia-Xun Li
- Department of Chemical Engineering and Biotechnology, Tatung University, Taipei 104, Taiwan
| | - Su-Chun How
- Department of Chemical Engineering and Biotechnology, Tatung University, Taipei 104, Taiwan.
| |
Collapse
|
2
|
Mahdavimehr M, Kaboudin B, Alaie S, Tondkar F, Eshkaftaki ZM, Ebrahim-Habibi MB, Ghashghaee M, Tahmasebi E, Zhang T, Gu Y, Meratan AA. Inhibition of cytotoxic self-assembly of HEWL through promoting fibrillation by new synthesized α-hydroxycarbamoylphosphinic acids. RSC Adv 2024; 14:31227-31242. [PMID: 39355328 PMCID: PMC11443501 DOI: 10.1039/d4ra02969k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/04/2024] [Indexed: 10/03/2024] Open
Abstract
The main objective of the present study is to investigate the potency of new synthesized hydroxycarbamoyl phosphinic acid derivatives in modulating cytotoxic fibrillogenesis of hen egg white lysozyme (HEWL), as a common model in protein aggregation studies. Hydroxycarbamoyl phosphinic acid derivatives were prepared by the reaction of α-hydroxyalkylphosphinic acids with isocyanates (or isothiocyanates) in the presence of trimethylsilyl chloride (TMSCl). The designed process involves the condensation reaction leading to formation of new C sp2-P bond formation. The synthesis and purity of novel designed compounds were confirmed by NMR, LC-MS, and HPLC techniques. A range of experiments, including thioflavin T (ThT) and 8-anilino-1-naphthalenesulfonic acid (ANS) fluorescence assays, Congo red binding measurement, atomic force microscopy imaging, MTT-based cell viability and hemolysis assays were employed to investigate anti-amyloidogenic effects of tested compounds. The obtained results demonstrate that these compounds are able to significantly modulate the self-assembly process of HEWL via shortening of nucleation phase leading to the acceleration of fibrillation and appearance of very large and thick fibrils with decreased surface hydrophobicity and cytotoxicity. Based on ANS binding data, we suggest that increased exposure of hydrophobic patches of oligomeric species is the possible mechanism by which tested compounds promote self-assembly process of HEWL. Fluorescence anisotropy and molecular docking studies indicate the interaction of both synthesized compounds with HEWL, and more specifically with residues that are situated in the highly aggregation-prone β-domain region of protein. This study unveils the potential of hydroxyalkylphosphinic acids as modulators of amyloid fibrillation highlighting these compounds as a promising approach for targeting protein aggregates associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohsen Mahdavimehr
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Babak Kaboudin
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Saied Alaie
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Farimah Tondkar
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Zahra Mahmoudi Eshkaftaki
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | | | - Mojtaba Ghashghaee
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Elham Tahmasebi
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Tianjian Zhang
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology Wuhan 430074 China
| | - Yanlong Gu
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology Wuhan 430074 China
| | - Ali Akbar Meratan
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| |
Collapse
|
3
|
Roy S, Srinivasan VR, Arunagiri S, Mishra N, Bhatia A, Shejale KP, Prajapati KP, Kar K, Anand BG. Molecular insights into the phase transition of lysozyme into amyloid nanostructures: Implications of therapeutic strategies in diverse pathological conditions. Adv Colloid Interface Sci 2024; 331:103205. [PMID: 38875805 DOI: 10.1016/j.cis.2024.103205] [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: 09/17/2023] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024]
Abstract
Lysozyme, a well-known bacteriolytic enzyme, exhibits a fascinating yet complex behavior when it comes to protein aggregation. Under certain conditions, this enzyme undergoes flexible transformation, transitioning from partially unfolded intermediate units of native conformers into complex cross-β-rich nano fibrillar amyloid architectures. Formation of such lysozyme amyloids has been implicated in a multitude of pathological and medical severities, like hepatic dysfunction, hepatomegaly, splenic rupture as well as spleen dysfunction, nephropathy, sicca syndrome, renal dysfunction, renal amyloidosis, and systemic amyloidosis. In this comprehensive review, we have attempted to provide in-depth insights into the aggregating behavior of lysozyme across a spectrum of variables, including concentrations, temperatures, pH levels, and mutations. Our objective is to elucidate the underlying mechanisms that govern lysozyme's aggregation process and to unravel the complex interplay between its structural attributes. Moreover, this work has critically examined the latest advancements in the field, focusing specifically on novel strategies and systems, that have been implemented to delay or inhibit the lysozyme amyloidogenesis. Apart from this, we have tried to explore and advance our fundamental understanding of the complex processes involved in lysozyme aggregation. This will help the research community to lay a robust foundation for screening, designing, and formulating targeted anti-amyloid therapeutics offering improved treatment modalities and interventions not only for lysozyme-linked amyloidopathy but for a wide range of amyloid-related disorders.
Collapse
Affiliation(s)
- Sindhujit Roy
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Venkat Ramanan Srinivasan
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Subash Arunagiri
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nishant Mishra
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Anubhuti Bhatia
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Kiran P Shejale
- Dept. of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, South Korea
| | - Kailash Prasad Prajapati
- Biophysical and Biomaterials Research Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Karunakar Kar
- Biophysical and Biomaterials Research Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India..
| | - Bibin Gnanadhason Anand
- Biomolecular Self-Assembly Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India..
| |
Collapse
|
4
|
Cardoso S, Carvalho C, Correia SC, Moreira PI. Protective effects of 2,4-dinitrophenol in okadaic acid-induced cellular model of Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167222. [PMID: 38729530 DOI: 10.1016/j.bbadis.2024.167222] [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: 02/02/2024] [Revised: 04/28/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Alzheimer's disease (AD) research started several decades ago and despite the many efforts employed to develop new treatments or approaches to slow and/or revert disease progression, AD treatment remains an unsolved issue. Knowing that mitochondria loss of function is a central hub for many AD-associated pathophysiological processes, there has been renewed interest in exploring mitochondria as targets for intervention. In this perspective, the present study was aimed to investigate the possible beneficial effects of 2,4 dinitrophenol (DNP), a mitochondrial uncoupler agent, in an in vitro model of AD. Retinoic acid-induced differentiated SH-SY5Y cells were incubated with okadaic acid (OA), a neurotoxin often used as an AD experimental model, and/or with DNP. OA caused a decrease in neuronal cells viability, induced multiple mitochondrial anomalies including increased levels of reactive oxygen species, decreased bioenergetics and mitochondria content markers, and an altered mitochondria morphology. OA-treated cells also presented increased lipid peroxidation levels, and overactivation of tau related kinases (GSK3β, ERK1/2 and AMPK) alongside with a significant augment in tau protein phosphorylation levels. Interestingly, DNP co-treatment ameliorated and rescued OA-induced detrimental effects not only on mitochondria but also but also reinstated signaling pathways homeostasis and ameliorated tau pathology. Overall, our results show for the first time that DNP has the potential to preserve mitochondria homeostasis under a toxic insult, like OA exposure, as well as to reestablish cellular signaling homeostasis. These observations foster the idea that DNP, as a mitochondrial modulator, might represent a new avenue for treatment of AD.
Collapse
Affiliation(s)
- Susana Cardoso
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; IIIU - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal.
| | - Cristina Carvalho
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; IIIU - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Sónia C Correia
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; IIIU - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Paula I Moreira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| |
Collapse
|
5
|
Tang J, Huang H, Muirhead RCJ, Zhou Y, Li J, DeFelice J, Kopanitsa MV, Serneels L, Davey K, Tilley BS, Gentleman S, Matthews PM. Associations of amyloid-β oligomers and plaques with neuropathology in the App NL-G-F mouse. Brain Commun 2024; 6:fcae218. [PMID: 39035420 PMCID: PMC11258573 DOI: 10.1093/braincomms/fcae218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 03/22/2024] [Accepted: 06/23/2024] [Indexed: 07/23/2024] Open
Abstract
Amyloid-β pathology and neurofibrillary tangles lead to glial activation and neurodegeneration in Alzheimer's disease. In this study, we investigated the relationships between the levels of amyloid-β oligomers, amyloid-β plaques, glial activation and markers related to neurodegeneration in the App NL-G-F triple mutation mouse line and in a knock-in line homozygous for the common human amyloid precursor protein (App hu mouse). The relationships between neuropathological features were characterized with immunohistochemistry and imaging mass cytometry. Markers assessing human amyloid-β proteins, microglial and astrocytic activation and neuronal and synaptic densities were used in mice between 2.5 and 12 months of age. We found that amyloid-β oligomers were abundant in the brains of App hu mice in the absence of classical amyloid-β plaques. These brains showed morphological changes consistent with astrocyte activation but no evidence of microglial activation or synaptic or neuronal pathology. In contrast, both high levels of amyloid-β oligomers and numerous plaques accumulated in App NL-G-F mice in association with substantial astrocytic and microglial activation. The increase in amyloid-β oligomers over time was more strongly correlated with astrocytic than with microglia activation. Spatial analyses suggested that activated microglia were more closely associated with amyloid-β oligomers than with amyloid-β plaques in App NL-G-F mice, which also showed age-dependent decreases in neuronal and synaptic density markers. A comparative study of the two models highlighted the dependence of glial and neuronal pathology on the nature and aggregation state of the amyloid-β peptide. Astrocyte activation and neuronal pathology appeared to be more strongly associated with amyloid-β oligomers than with amyloid-β plaques, although amyloid-β plaques were associated with microglia activation.
Collapse
Affiliation(s)
- Jiabin Tang
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Department of Anesthesiology, Weill Cornell Medicine, Cornell University, New York, NY 11106, USA
| | - Helen Huang
- Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Robert C J Muirhead
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Randall Centre for Cell and Molecular Biophysics, Kings College London, London SE5 9RX, UK
| | - Yue Zhou
- Department of Mechanical Engineering, Roberts Engineering Building, University College London, London WC1E 7JE, UK
| | - Junheng Li
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
| | - John DeFelice
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Maksym V Kopanitsa
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Lutgarde Serneels
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), 9052 Gent, Belgium
| | - Karen Davey
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- UK Dementia Research Institute, Kings College London, Denmark Hill Campus, London SE5 9RX, UK
| | - Bension S Tilley
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Steve Gentleman
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Paul M Matthews
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| |
Collapse
|
6
|
Fan J, Liang L, Zhou X, Ouyang Z. Accelerating protein aggregation and amyloid fibrillation for rapid inhibitor screening. Chem Sci 2024; 15:6853-6859. [PMID: 38725489 PMCID: PMC11077537 DOI: 10.1039/d4sc00437j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/04/2024] [Indexed: 05/12/2024] Open
Abstract
The accumulation and deposition of amyloid fibrils, also known as amyloidosis, in tissues and organs of patients has been found to be linked to numerous devastating neurodegenerative diseases. The aggregation of proteins to form amyloid fibrils, however, is a slow pathogenic process, and is a major issue for the evaluation of the effectiveness of inhibitors in new drug discovery and screening. Here, we used microdroplet reaction technology to accelerate the amyloid fibrillation process, monitored the process to shed light on the fundamental mechanism of amyloid self-assembly, and demonstrated the value of the technology in the rapid screening of potential inhibitor drugs. Proteins in microdroplets accelerated to form fibrils in milliseconds, enabling an entire cycle of inhibitor screening for Aβ40 within 3 minutes. The technology would be of broad interest to drug discovery and therapeutic design to develop treatments for diseases associated with protein aggregation and fibrillation.
Collapse
Affiliation(s)
- Jingjin Fan
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University Beijing 100084 China
| | - Liwen Liang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University Beijing 100084 China
| | - Xiaoyu Zhou
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University Beijing 100084 China
| | - Zheng Ouyang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University Beijing 100084 China
| |
Collapse
|
7
|
Hyperoside alleviates toxicity of β-amyloid via endoplasmic reticulum-mitochondrial calcium signal transduction cascade in APP/PS1 double transgenic Alzheimer's disease mice. Redox Biol 2023; 61:102637. [PMID: 36821955 PMCID: PMC9975698 DOI: 10.1016/j.redox.2023.102637] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized by a decline in cognitive function. The β-amyloid (Aβ) hypothesis suggests that Aβ peptides can spontaneously aggregate into β-fragment-containing oligomers and protofibrils, and this activation of the amyloid pathway alters Ca2+ signaling in neurons, leading to neurotoxicity and thus apoptosis of neuronal cells. In our study, a blood-brain barrier crossing flavonol glycoside hyperoside was identified with anti-Aβ aggregation, BACE inhibitory, and neuroprotective effect in cellular or APP/PSEN1 double transgenic Alzheimer's disease mice model. While our pharmacokinetic data confirmed that intranasal administration of hyperoside resulted in a higher bio-availability in mice brain, further in vivo studies revealed that it improved motor deficit, spatial memory and learning ability of APP/PSEN1 mice with reducing level of Aβ plaques and GFAP in the cortex and hippocampus. Bioinformatics, computational docking and in vitro assay results suggested that hyperoside bind to Aβ and interacted with ryanodine receptors, then regulated cellular apoptosis via endoplasmic reticulum-mitochondrial calcium (Ca2+) signaling pathway. Consistently, it was confirmed that hyperoside increased Bcl2, decreased Bax and cyto-c protein levels, and ameliorated neuronal cell death in both in vitro and in vivo model. By regulating Aβ-induced cell death via regulation on Ca2+ signaling cascade and mitochondrial membrane potential, our study suggested that hyperoside may work as a potential therapeutic agent or preventive remedy for Alzheimer's disease.
Collapse
|
8
|
Screening the extraction process of phenolic compounds from pressed grape seed residue: Towards an integrated and sustainable management of viticultural waste. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
9
|
Using Sugar-Derived Nanoparticles to Mitigate Amyloid Fibril Formation of Lysozyme. J Taiwan Inst Chem Eng 2022. [DOI: 10.1016/j.jtice.2022.104360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
10
|
Basu A, Mahammad A, Das A. Inhibition of the formation of lysozyme fibrillar assemblies by the isoquinoline alkaloid coralyne. NEW J CHEM 2022. [DOI: 10.1039/d1nj06007d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The isoquinoline alkaloid coralyne can efficiently attenuate fibrillogenesis in lysozyme.
Collapse
Affiliation(s)
- Anirban Basu
- Department of Chemistry, Vidyasagar University, Midnapore 721 102, India
| | - Adil Mahammad
- Department of Chemistry, Vidyasagar University, Midnapore 721 102, India
| | - Arindam Das
- Department of Chemistry, Vidyasagar University, Midnapore 721 102, India
| |
Collapse
|
11
|
Raut S, Patel R, Pervaiz I, Al-Ahmad AJ. Abeta Peptides Disrupt the Barrier Integrity and Glucose Metabolism of Human Induced Pluripotent Stem Cell-Derived Brain Microvascular Endothelial Cells. Neurotoxicology 2022; 89:110-120. [DOI: 10.1016/j.neuro.2022.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022]
|
12
|
Ashrafian H, Zadeh EH, Tajbakhsh M, Majid N, Srivastava GN, Khan RH. Discovery of a tetracyclic indole alkaloid that postpones fibrillation of hen egg white lysozyme protein. Int J Biol Macromol 2021; 183:1939-1947. [PMID: 34097957 DOI: 10.1016/j.ijbiomac.2021.05.212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/11/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
Protein aggregation, such as amyloid fibril formation, is molecular hallmark of many neurodegenerative disorders including Alzheimer's, Parkinson's, and Prion disease. Indole alkaloids are well-known as the compounds having the ability to inhibit protein fibrillation. In this study, we experimentally and computationally have investigated the anti-amyloid property of a derivative of a synthesized tetracyclic indole alkaloid (TCIA), possessing capable functional groups. The fibrillation reaction of Hen White Egg Lysozyme (HEWL) was performed in absence and presence of the indole alkaloid. For quantitative analysis, we used Thioflovin T binding assay which showed ~50% reduction in fibril formation in the presence of 20 μM TCIA. Using TEM imaging, we observed a significant morphological change in our model protein in the presence of TCIA. In addition, we exploited FT-IR assay by which Amide I peak's shifting toward lower wavenumber was clearly observed. Using Molecular Docking, the interaction of the inhibitor (TCIA) with the protein's amyloidogenic region was modeled. Also, different biophysical parameters were calculated by Molecular Dynamics (MD) simulation. Various biochemical assays, conformational change, and hydrophobicity exposure of the protein during amyloid formation indicated that the compound assists HEWL to keep its native structure via destabilizing β-sheet structure.
Collapse
Affiliation(s)
- Hossein Ashrafian
- Department of Chemistry and Biochemistry, the Ohio State University, Columbus, OH, USA; Biochemistry Lab, Chemistry department, Sharif University of Technology, Tehran, Iran.
| | | | | | - Nabeela Majid
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202 002, India
| | - Gopal N Srivastava
- Department of Chemistry and Biochemistry, the Ohio State University, Columbus, OH, USA
| | - Rizwan Hassan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202 002, India.
| |
Collapse
|
13
|
Pignataro P, Dicarlo M, Zerlotin R, Zecca C, Dell’Abate MT, Buccoliero C, Logroscino G, Colucci S, Grano M. FNDC5/Irisin System in Neuroinflammation and Neurodegenerative Diseases: Update and Novel Perspective. Int J Mol Sci 2021; 22:ijms22041605. [PMID: 33562601 PMCID: PMC7915567 DOI: 10.3390/ijms22041605] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Irisin, the circulating peptide originating from fibronectin type III domain-containing protein 5 (FNDC5), is mainly expressed by muscle fibers under peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) control during exercise. In addition to several beneficial effects on health, physical activity positively affects nervous system functioning, particularly the hippocampus, resulting in amelioration of cognition impairments. Recently, FNDC5/irisin detection in hippocampal neurons and the presence of irisin in the cerebrospinal fluid opened a new intriguing chapter in irisin history. Interestingly, in the hippocampus of mice, exercise increases FNDC5 levels and upregulates brain-derived neurotrophic factor (BDNF) expression. BDNF, displaying neuroprotection and anti-inflammatory effects, is mainly produced by microglia and astrocytes. In this review, we discuss how these glial cells can morphologically and functionally switch during neuroinflammation by modulating the expression of a plethora of neuroprotective or neurotoxic factors. We also focus on studies investigating the irisin role in neurodegenerative diseases (ND). The emerging involvement of irisin as a mediator of the multiple positive effects of exercise on the brain needs further studies to better deepen this issue and the potential use in therapeutic approaches for neuroinflammation and ND.
Collapse
Affiliation(s)
- Patrizia Pignataro
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy; (P.P.); (M.D.); (G.L.); (S.C.)
- Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.Z.); (C.B.)
| | - Manuela Dicarlo
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy; (P.P.); (M.D.); (G.L.); (S.C.)
| | - Roberta Zerlotin
- Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.Z.); (C.B.)
| | - Chiara Zecca
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari, “Pia Fondazione Card G. Panico” Hospital Tricase, 73039 Lecce, Italy; (C.Z.); (M.T.D.)
| | - Maria Teresa Dell’Abate
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari, “Pia Fondazione Card G. Panico” Hospital Tricase, 73039 Lecce, Italy; (C.Z.); (M.T.D.)
| | - Cinzia Buccoliero
- Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.Z.); (C.B.)
| | - Giancarlo Logroscino
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy; (P.P.); (M.D.); (G.L.); (S.C.)
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari, “Pia Fondazione Card G. Panico” Hospital Tricase, 73039 Lecce, Italy; (C.Z.); (M.T.D.)
| | - Silvia Colucci
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy; (P.P.); (M.D.); (G.L.); (S.C.)
| | - Maria Grano
- Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.Z.); (C.B.)
- Correspondence: ; Tel.: +39-080-5478-361
| |
Collapse
|
14
|
Mahmoudinobar F, Nilsson BL, Dias CL. Effects of Ions and Small Compounds on the Structure of Aβ 42 Monomers. J Phys Chem B 2021; 125:1085-1097. [PMID: 33481611 DOI: 10.1021/acs.jpcb.0c09617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Aggregation of amyloid-β (Aβ) proteins in the brain is a hallmark of Alzheimer's disease. This phenomenon can be promoted or inhibited by adding small molecules to the solution where Aβ is embedded. These molecules affect the ensemble of conformations sampled by Aβ monomers even before aggregation starts. Here, we perform extensive all-atom replica exchange molecular dynamics (REMD) simulations to provide a comparative study of the ensemble of conformations sampled by Aβ42 monomers in solutions that promote (i.e., aqueous solution containing NaCl) and inhibit (i.e., aqueous solutions containing scyllo-inositol or 4-aminophenol) aggregation. Simulations performed in pure water are used as our reference. We find that secondary-structure content is only affected in an antagonistic manner by promoters and inhibitors at the C-terminus and the central hydrophilic core. Moreover, the end of the C-terminus binds more favorably to the central hydrophobic core region of Aβ42 in NaCl adopting a type of strand-loop-strand structure that is disfavored by inhibitors. Nonpolar residues that form the dry core of larger aggregates of Aβ42 (e.g., PDB ID 2BEG) are found at close proximity in these strand-loop-strand structures, suggesting that their formation could play an important role in initiating nucleation. In the presence of inhibitors, the C-terminus binds the central hydrophilic core with a higher probability than in our reference simulation. This sensitivity of the C-terminus, which is affected in an antagonistic manner by inhibitors and promoters, provides evidence for its critical role in accounting for aggregation.
Collapse
Affiliation(s)
- Farbod Mahmoudinobar
- Department of Physics, New Jersey Institute of Technology, Newark, New Jersey 07102-1982, United States
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, New York 14627, United States
| | - Cristiano L Dias
- Department of Physics, New Jersey Institute of Technology, Newark, New Jersey 07102-1982, United States
| |
Collapse
|
15
|
Kim M, Kang J, Lee M, Han J, Nam G, Tak E, Kim MS, Lee HJ, Nam E, Park J, Oh SJ, Lee JY, Lee JY, Baik MH, Lim MH. Minimalistic Principles for Designing Small Molecules with Multiple Reactivities against Pathological Factors in Dementia. J Am Chem Soc 2020; 142:8183-8193. [PMID: 32233474 DOI: 10.1021/jacs.9b13100] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multiple pathogenic elements, including reactive oxygen species, amyloidogenic proteins, and metal ions, are associated with the development of neurodegenerative disorders. We report minimalistic redox-based principles for preparing compact aromatic compounds by derivatizing the phenylene moiety with various functional groups. These molecular agents display enhanced reactivities against multiple targets such as free radicals, metal-free amyloid-β (Aβ), and metal-bound Aβ that are implicated in the most common form of dementia, Alzheimer's disease (AD). Mechanistic studies reveal that the redox properties of these reagents are essential for their function. Specifically, they engage in oxidative reactions with metal-free and metal-bound Aβ, leading to chemical modifications of the Aβ peptides to form covalent adducts that alter the aggregation of Aβ. Moreover, the administration of the most promising candidate significantly attenuates the amyloid pathology in the brains of AD transgenic mice and improves their cognitive defects. Our studies demonstrate an efficient and effective redox-based strategy for incorporating multiple functions into simple molecular reagents.
Collapse
Affiliation(s)
- Mingeun Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Juhye Kang
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Misun Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.,Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jiyeon Han
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Geewoo Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eunyoung Tak
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Min Sun Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University, Gongju 32588, Republic of Korea
| | - Eunju Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jiyong Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.,Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Soo Jin Oh
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Ji-Yoon Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Joo-Yong Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Mu-Hyun Baik
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.,Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
16
|
Salahuddin P, Khan RH, Uversky VN. Comprehensive analysis of the molecular docking of small molecule inhibitors to the Aβ1–40peptide and its Osaka-mutant: insights into the molecular mechanisms of Aβ-peptide inhibition. J Biomol Struct Dyn 2019; 38:4536-4566. [DOI: 10.1080/07391102.2019.1697367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Parveen Salahuddin
- DISC, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Vladimir N. Uversky
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Russia
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
17
|
Bodart-Santos V, de Carvalho LRP, de Godoy MA, Batista AF, Saraiva LM, Lima LG, Abreu CA, De Felice FG, Galina A, Mendez-Otero R, Ferreira ST. Extracellular vesicles derived from human Wharton's jelly mesenchymal stem cells protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. Stem Cell Res Ther 2019; 10:332. [PMID: 31747944 PMCID: PMC6864996 DOI: 10.1186/s13287-019-1432-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/26/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been explored as promising tools for treatment of several neurological and neurodegenerative diseases. MSCs release abundant extracellular vesicles (EVs) containing a variety of biomolecules, including mRNAs, miRNAs, and proteins. We hypothesized that EVs derived from human Wharton’s jelly would act as mediators of the communication between hMSCs and neurons and could protect hippocampal neurons from damage induced by Alzheimer’s disease-linked amyloid beta oligomers (AβOs). Methods We isolated and characterized EVs released by human Wharton’s jelly mesenchymal stem cells (hMSC-EVs). The neuroprotective action of hMSC-EVs was investigated in primary hippocampal cultures exposed to AβOs. Results hMSC-EVs were internalized by hippocampal cells in culture, and this was enhanced in the presence of AβOs in the medium. hMSC-EVs protected hippocampal neurons from oxidative stress and synapse damage induced by AβOs. Neuroprotection by hMSC-EVs was mediated by catalase and was abolished in the presence of the catalase inhibitor, aminotriazole. Conclusions hMSC-EVs protected hippocampal neurons from damage induced by AβOs, and this was related to the transfer of enzymatically active catalase contained in EVs. Results suggest that hMSC-EVs should be further explored as a cell-free therapeutic approach to prevent neuronal damage in Alzheimer’s disease.
Collapse
Affiliation(s)
- Victor Bodart-Santos
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Luiza R P de Carvalho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Mariana A de Godoy
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - André F Batista
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Leonardo M Saraiva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Luize G Lima
- National Cancer Institute, Rio de Janeiro, RJ, 20230-240, Brazil
| | - Carla Andreia Abreu
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.,Centre for Neuroscience Studies and Department of Psychiatry, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Antonio Galina
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Rosalia Mendez-Otero
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, 21941-590, Brazil.
| | - Sergio T Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil. .,Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
18
|
Owen MC, Gnutt D, Gao M, Wärmländer SKTS, Jarvet J, Gräslund A, Winter R, Ebbinghaus S, Strodel B. Effects of in vivo conditions on amyloid aggregation. Chem Soc Rev 2019; 48:3946-3996. [PMID: 31192324 DOI: 10.1039/c8cs00034d] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the grand challenges of biophysical chemistry is to understand the principles that govern protein misfolding and aggregation, which is a highly complex process that is sensitive to initial conditions, operates on a huge range of length- and timescales, and has products that range from protein dimers to macroscopic amyloid fibrils. Aberrant aggregation is associated with more than 25 diseases, which include Alzheimer's, Parkinson's, Huntington's, and type II diabetes. Amyloid aggregation has been extensively studied in the test tube, therefore under conditions that are far from physiological relevance. Hence, there is dire need to extend these investigations to in vivo conditions where amyloid formation is affected by a myriad of biochemical interactions. As a hallmark of neurodegenerative diseases, these interactions need to be understood in detail to develop novel therapeutic interventions, as millions of people globally suffer from neurodegenerative disorders and type II diabetes. The aim of this review is to document the progress in the research on amyloid formation from a physicochemical perspective with a special focus on the physiological factors influencing the aggregation of the amyloid-β peptide, the islet amyloid polypeptide, α-synuclein, and the hungingtin protein.
Collapse
Affiliation(s)
- Michael C Owen
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic
| | - David Gnutt
- Institute of Physical and Theoretical Chemistry, TU Braunschweig, Rebenring 56, 38106 Braunschweig, Germany and Lead Discovery Wuppertal, Bayer AG, 42096 Wuppertal, Germany
| | - Mimi Gao
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn Str. 4a, 44227 Dortmund, Germany and Sanofi-Aventis Deutschland GmbH, R&D, Industriepark Höchst, 65926 Frankfurt, Germany
| | - Sebastian K T S Wärmländer
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 106 91 Stockholm, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 106 91 Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 106 91 Stockholm, Sweden
| | - Roland Winter
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn Str. 4a, 44227 Dortmund, Germany
| | - Simon Ebbinghaus
- Institute of Physical and Theoretical Chemistry, TU Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry, Forschungszentrum Jülich, 42525 Jülich, Germany. and Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
19
|
Moussa-Pacha NM, Abdin SM, Omar HA, Alniss H, Al-Tel TH. BACE1 inhibitors: Current status and future directions in treating Alzheimer's disease. Med Res Rev 2019; 40:339-384. [PMID: 31347728 DOI: 10.1002/med.21622] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/22/2019] [Accepted: 06/13/2019] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is an irreversible, progressive neurodegenerative brain disorder with no current cure. One of the important therapeutic approaches of AD is the inhibition of β-site APP cleaving enzyme-1 (BACE1), which is involved in the rate-limiting step of the cleavage process of the amyloid precursor protein (APP) leading to the generation of the neurotoxic amyloid β (Aβ) protein after the γ-secretase completes its function. The produced insoluble Aβ aggregates lead to plaques deposition and neurodegeneration. BACE1 is, therefore, one of the attractive targets for the treatment of AD. This approach led to the development of potent BACE1 inhibitors, many of which were advanced to late stages in clinical trials. Nonetheless, the high failure rate of lead drug candidates targeting BACE1 brought to the forefront the need for finding new targets to uncover the mystery behind AD. In this review, we aim to discuss the most promising classes of BACE1 inhibitors with a description and analysis of their pharmacodynamic and pharmacokinetic parameters, with more focus on the lead drug candidates that reached late stages of clinical trials, such as MK8931, AZD-3293, JNJ-54861911, E2609, and CNP520. In addition, the manuscript discusses the safety concerns and insignificant physiological effects, which were highlighted for the most successful BACE1 inhibitors. Furthermore, the review demonstrates with increasing evidence that despite tremendous efforts and promising results conceived with BACE1 inhibitors, the latest studies suggest that their clinical use for treating Alzheimer's disease should be reconsidered. Finally, the review sheds light on alternative therapeutic options for targeting AD.
Collapse
Affiliation(s)
- Nour M Moussa-Pacha
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Shifaa M Abdin
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy and College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hasan Alniss
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy and College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Taleb H Al-Tel
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy and College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
20
|
How SC, Hsin A, Chen GY, Hsu WT, Yang SM, Chou WL, Chou SH, Wang SSS. Exploring the influence of brilliant blue G on amyloid fibril formation of lysozyme. Int J Biol Macromol 2019; 138:37-48. [PMID: 31295491 DOI: 10.1016/j.ijbiomac.2019.07.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/28/2019] [Accepted: 07/07/2019] [Indexed: 12/19/2022]
Abstract
Evidence suggests that amyloid fibril mitigation/inhibition is considered a promising approach toward treating amyloid diseases. In this work, we first examined how amyloid fibrillogenesis of lysozyme was affected by BBG, a safe triphenylmethane compound with nice blood-brain-barrier-permeability, and found that shorter fibrillar species were formed in the lysozyme samples treated with BBG. Next, alterations in the features including the secondary as well as tertiary structure, extent of aggregation, and molecular distribution of lysozyme triggered by the addition of BBG were examined by various spectroscopic techniques, right-angle light scattering, dynamic light scattering, and SDS-PAGE. In addition, we have investigated how BBG affected the lysozyme fibril-induced cytotoxicity in SH-SY5Y cells. We found that a large quantity of shorter fibrillar species and more lysozyme monomers were present in the samples treated with BBG. Also, the addition of BBG rescued SH-SY5Y cells from cell death induced by amyloid fibrils of lysozyme. Finally, information about the binding sites and interacting forces involved in the BBG-lysozyme interaction was further explored using synchronous fluorescence and molecular docking approaches. Molecular docking results revealed that, apart from the hydrophobic interaction(s), hydrogen bonding, electrostatic interactions, and van der Waal forces may also be involved in the binding interaction.
Collapse
Affiliation(s)
- Su-Chun How
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Ai Hsin
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Guan-Yu Chen
- Department of Life Science, Fu-Jen Catholic University, Xinzhuang Dist., New Taipei City, Taiwan
| | - Wei-Tse Hsu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Szu-Ming Yang
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Wei-Lung Chou
- Department of Safety, Health and Environmental Engineering, Hungkuang University, Sha Lu, Taichung City 433, Taiwan.
| | - Shiu-Huey Chou
- Department of Life Science, Fu-Jen Catholic University, Xinzhuang Dist., New Taipei City, Taiwan.
| | - Steven S-S Wang
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
21
|
Khan RH, Siddiqi MK, Uversky VN, Salahuddin P. Molecular docking of Aβ1–40 peptide and its Iowa D23N mutant using small molecule inhibitors: Possible mechanisms of Aβ-peptide inhibition. Int J Biol Macromol 2019; 127:250-270. [DOI: 10.1016/j.ijbiomac.2018.12.271] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/21/2018] [Accepted: 12/31/2018] [Indexed: 12/11/2022]
|
22
|
Geisler JG. 2,4 Dinitrophenol as Medicine. Cells 2019; 8:cells8030280. [PMID: 30909602 PMCID: PMC6468406 DOI: 10.3390/cells8030280] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022] Open
Abstract
In the sanctity of pure drug discovery, objective reasoning can become clouded when pursuing ideas that appear unorthodox, but are spot on physiologically. To put this into historical perspective, it was an unorthodox idea in the 1950’s to suggest that warfarin, a rat poison, could be repositioned into a breakthrough drug in humans to protect against strokes as a blood thinner. Yet it was approved in 1954 as Coumadin® and has been prescribed to billions of patients as a standard of care. Similarly, no one can forget the horrific effects of thalidomide, prescribed or available without a prescription, as both a sleeping pill and “morning sickness” anti-nausea medication targeting pregnant women in the 1950’s. The “thalidomide babies” became the case-in-point for the need of strict guidelines by the U.S. Food & Drug Administration (FDA) or full multi-species teratogenicity testing before drug approval. More recently it was found that thalidomide is useful in graft versus host disease, leprosy and resistant tuberculosis treatment, and as an anti-angiogenesis agent as a breakthrough drug for multiple myeloma (except for pregnant female patients). Decades of diabetes drug discovery research has historically focused on every possible angle, except, the energy-out side of the equation, namely, raising mitochondrial energy expenditure with chemical uncouplers. The idea of “social responsibility” allowed energy-in agents to be explored and the portfolio is robust with medicines of insulin sensitizers, insulin analogues, secretagogues, SGLT2 inhibitors, etc., but not energy-out medicines. The primary reason? It appeared unorthodox, to return to exploring a drug platform used in the 1930s in over 100,000 obese patients used for weight loss. This is over 80-years ago and prior to Dr Peter Mitchell explaining the mechanism of how mitochondrial uncouplers, like 2,4-dinitrophenol (DNP) even worked by three decades later in 1961. Although there is a clear application for metabolic disease, it was not until recently that this platform was explored for its merit at very low, weight-neutral doses, for treating insidious human illnesses and completely unrelated to weight reduction. It is known that mitochondrial uncouplers specifically target the entire organelle’s physiology non-genomically. It has been known for years that many neuromuscular and neurodegenerative diseases are associated with overt production of reactive oxygen species (ROSs), a rise in isoprostanes (biomarker of mitochondrial ROSs in urine or blood) and poor calcium (Ca2+) handing. It has also been known that mitochondrial uncouplers lower ROS production and Ca2+ overload. There is evidence that elevation of isoprostanes precedes disease onset, in Alzheimer’s Disease (AD). It is also curious, why so many neurodegenerative diseases of known and unknown etiology start at mid-life or later, such as Multiple Sclerosis (MS), Huntington Disease (HD), AD, Parkinson Disease, and Amyotrophic Lateral Sclerosis (ALS). Is there a relationship to a buildup of mutations that are sequestered over time due to ROSs exceeding the rate of repair? If ROS production were managed, could disease onset due to aging be delayed or prevented? Is it possible that most, if not all neurodegenerative diseases are manifested through mitochondrial dysfunction? Although DNP, a historic mitochondrial uncoupler, was used in the 1930s at high doses for obesity in well over 100,000 humans, and so far, it has never been an FDA-approved drug. This review will focus on the application of using DNP, but now, repositioned as a potential disease-modifying drug for a legion of insidious diseases at much lower and paradoxically, weight neutral doses. DNP will be addressed as a treatment for “metabesity”, an emerging term related to the global comorbidities associated with the over-nutritional phenotype; obesity, diabetes, nonalcoholic steatohepatitis (NASH), metabolic syndrome, cardiovascular disease, but including neurodegenerative disorders and accelerated aging. Some unexpected drug findings will be discussed, such as DNP’s induction of neurotrophic growth factors involved in neuronal heath, learning and cognition. For the first time in 80’s years, the FDA has granted (to Mitochon Pharmaceutical, Inc., Blue Bell, PA, USA) an open Investigational New Drug (IND) approval to begin rigorous clinical testing of DNP for safety and tolerability, including for the first ever, pharmacokinetic profiling in humans. Successful completion of Phase I clinical trial will open the door to explore the merits of DNP as a possible treatment of people with many truly unmet medical needs, including those suffering from HD, MS, PD, AD, ALS, Duchenne Muscular Dystrophy (DMD), and Traumatic Brain Injury (TBI).
Collapse
Affiliation(s)
- John G Geisler
- Mitochon Pharmaceuticals, Inc., 970 Cross Lane, Blue Bell, PA 19422, USA.
| |
Collapse
|
23
|
Yoon JH, Lee YS, Kim O, Ashktorab H, Smoot DT, Nam SW, Park WS. NKX6.3 protects against gastric mucosal atrophy by downregulating β-amyloid production. World J Gastroenterol 2019; 25:330-345. [PMID: 30686901 PMCID: PMC6343100 DOI: 10.3748/wjg.v25.i3.330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Atrophic gastritis is characterized by loss of appropriate glands and reduction in gastric secretory function due to chronic inflammatory processes in gastric mucosa. Moreover, atrophic gastritis is considered as a precancerous condition of gastric cancer. However, little is known about the molecular mechanism underlying gastric mucosal atrophy and its contribution to gastric carcinogenesis. Thus, we hypothesized that transcription factor NKX6.3 might be involved in maintaining gastric epithelial homeostasis by regulating amyloid β (Aβ) production. AIM To determine whether NKX6.3 might protect against gastric mucosal atrophy by regulating Aβ production. METHODS We identified NKX6.3 depletion induced cell death by cell count and Western blot assay. Production and mechanism of Aβ oligomer were analyzed by enzyme-linked immunosorbent assay, Western blot, immunoprecipitation, real-time quantitative polymerase chain reaction and immunofluorescence analysis. We further validated the correlation between expression of NKX6.3, Helicobacter pylori CagA, Aβ oligomer, apolipoprotein E (ApoE), and β-secretase 1 (Bace1) in 55 gastric mucosae. RESULTS NKX6.3 depletion increased both adherent and floating cell populations in HFE-145 cells. Expression levels of cleaved caspase-3, -9, and poly ADP ribose polymerase were elevated in floating HFE-145shNKX6.3 cells. NKX6.3 depletion produced Aβ peptide oligomers, and increased expression of ApoE, amyloid precursor protein, Aβ, Bace1, low-density lipoprotein receptor, nicastrin, high mobility group box1, and receptor for advanced glycosylation end product proteins. In immunoprecipitation assay, γ-secretase complex was stably formed only in HFE-145shNKX6.3 cells. In gastric mucosae with atrophy, expression of Aβ peptide oligomer, ApoE, and Bace1 was detected and inversely correlated with NKX6.3 expression. Treatment with recombinant Aβ 1-42 produced Aβ oligomeric forms and decreased cell viability in HFE-145shNKX6.3 cells. Additionally, NKX6.3 depletion increased expression of inflammatory cytokines and cyclooxygenase-2. CONCLUSION NKX6.3 inhibits gastric mucosal atrophy by regulating Aβ accumulation and inflammatory reaction in gastric epithelial cells.
Collapse
Affiliation(s)
- Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Yeon Soo Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Olga Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Hassan Ashktorab
- Department of Medicine, Howard University, Washington, DC 20060, United States
| | - Duane T Smoot
- Department of Medicine, Meharry Medical Center, Nashville, TN 37208, United States
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| |
Collapse
|
24
|
Analysis of the interaction of para-sulfonatocalix[8]arene with free amino acids and a six residue segment of β-amyloid peptide as a potential treatment for Alzheimer’s disease. J INCL PHENOM MACRO 2019. [DOI: 10.1007/s10847-018-00879-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
25
|
Morsy A, Trippier PC. Current and Emerging Pharmacological Targets for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2019; 72:S145-S176. [PMID: 31594236 DOI: 10.3233/jad-190744] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
No cure or disease-modifying therapy for Alzheimer's disease (AD) has yet been realized. However, a multitude of pharmacological targets have been identified for possible engagement to enable drug discovery efforts for AD. Herein, we review these targets comprised around three main therapeutic strategies. First is an approach that targets the main pathological hallmarks of AD: amyloid-β (Aβ) oligomers and hyperphosphorylated tau tangles which primarily focuses on reducing formation and aggregation, and/or inducing their clearance. Second is a strategy that modulates neurotransmitter signaling. Comprising this strategy are the cholinesterase inhibitors and N-methyl-D-aspartate receptor blockade treatments that are clinically approved for the symptomatic treatment of AD. Additional targets that aim to stabilize neuron signaling through modulation of neurotransmitters and their receptors are also discussed. Finally, the third approach comprises a collection of 'sensitive targets' that indirectly influence Aβ or tau accumulation. These targets are proteins that upon Aβ accumulation in the brain or direct Aβ-target interaction, a modification in the target's function is induced. The process occurs early in disease progression, ultimately causing neuronal dysfunction. This strategy aims to restore normal target function to alleviate Aβ-induced toxicity in neurons. Overall, we generally limit our analysis to targets that have emerged in the last decade and targets that have been validated using small molecules in in vitro and/or in vivo models. This review is not an exhaustive list of all possible targets for AD but serves to highlight the most promising and critical targets suitable for small molecule drug intervention.
Collapse
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
26
|
SLOH, a carbazole-based fluorophore, mitigates neuropathology and behavioral impairment in the triple-transgenic mouse model of Alzheimer's disease. Neuropharmacology 2018; 131:351-363. [PMID: 29309769 DOI: 10.1016/j.neuropharm.2018.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/08/2017] [Accepted: 01/02/2018] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative dysfunction characterized by memory impairment and brings a heavy burden to old people both in developing and developed countries. Amyloid hypothesis reveals that aggregation and deposition of amyloid plaques are the cause of AD neurodegeneration. SLOH, a carbazole-based fluorophore, is reported to inhibit amyloid beta (Aβ) aggregation in vitro. In the current study, we intended to evaluate the protective effect of SLOH in a triple transgenic AD mouse model (3xTg-AD). 3xTg-AD (10-month-old) were treated with SLOH (0.5, 1 and 2 mg kg-1) for one month via intraperitoneal injection. After treatment, cognitive function was assessed by Morris Water Maze (MWM) and Y-maze tasks. In addition, biochemical estimations were used to examine the degree of Aβ deposition, tau hyperphosphorylation and neuroinflammation in the brains of 3xTg-AD mice. An in vitro study was conducted on human neuroblastoma (SH-SY5Y) cells to determine the activity of SLOH on tau and GSK-3β using western blot and immunofluorescence staining. One month treatment with SLOH significantly ameliorated memory impairments in 3xTg-AD mice in MWM and Y-maze tests. Moreover, SLOH treatment mitigated the level of amyloid plaques, tau hyperphosphorylation and neuroinflammation in the mouse brain. SLOH also reduced tau hyperphosphorylation and down-regulated GSK-3β activity in Aβ induced neurotoxic SH-SY5Y cells. The promising results in mitigating amyloid plaques, tau hyperphosphorylation, neuroinflammation and ameliorating cognitive deficits following one-month treatment suggest that SLOH could be a potential multi-target molecule for the AD treatment.
Collapse
|
27
|
de Godoy MA, Saraiva LM, de Carvalho LRP, Vasconcelos-Dos-Santos A, Beiral HJV, Ramos AB, Silva LRDP, Leal RB, Monteiro VHS, Braga CV, de Araujo-Silva CA, Sinis LC, Bodart-Santos V, Kasai-Brunswick TH, Alcantara CDL, Lima APCA, da Cunha-E Silva NL, Galina A, Vieyra A, De Felice FG, Mendez-Otero R, Ferreira ST. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. J Biol Chem 2017; 293:1957-1975. [PMID: 29284679 DOI: 10.1074/jbc.m117.807180] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a disabling and highly prevalent neurodegenerative condition, for which there are no effective therapies. Soluble oligomers of the amyloid-β peptide (AβOs) are thought to be proximal neurotoxins involved in early neuronal oxidative stress and synapse damage, ultimately leading to neurodegeneration and memory impairment in AD. The aim of the current study was to evaluate the neuroprotective potential of mesenchymal stem cells (MSCs) against the deleterious impact of AβOs on hippocampal neurons. To this end, we established transwell cocultures of rat hippocampal neurons and MSCs. We show that MSCs and MSC-derived extracellular vesicles protect neurons against AβO-induced oxidative stress and synapse damage, revealed by loss of pre- and postsynaptic markers. Protection by MSCs entails three complementary mechanisms: 1) internalization and degradation of AβOs; 2) release of extracellular vesicles containing active catalase; and 3) selective secretion of interleukin-6, interleukin-10, and vascular endothelial growth factor to the medium. Results support the notion that MSCs may represent a promising alternative for cell-based therapies in AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Renata B Leal
- From the Institute of Biophysics Carlos Chagas Filho
| | | | | | | | | | | | | | | | | | | | - Antonio Galina
- the Institute of Medical Biochemistry Leopoldo de Meis, and
| | - Adalberto Vieyra
- From the Institute of Biophysics Carlos Chagas Filho.,the National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | | | | | - Sergio T Ferreira
- From the Institute of Biophysics Carlos Chagas Filho, .,the Institute of Medical Biochemistry Leopoldo de Meis, and
| |
Collapse
|
28
|
Haribabu J, Ranade DS, Bhuvanesh NSP, Kulkarni PP, Karvembu R. Ru(II)-p
-cymene Thiosemicarbazone Complexes as Inhibitors of Amyloid β (Aβ) Peptide Aggregation and Aβ-Induced Cytotoxicity. ChemistrySelect 2017. [DOI: 10.1002/slct.201702390] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Jebiti Haribabu
- Department of Chemistry; National Institute of Technology; Tiruchirappalli 620015 India
| | - Dnyanesh S. Ranade
- Bioprospecting Group; Agharkar Research Institute; G. G. Agarkar Road Pune 411004 India
| | | | - Prasad P. Kulkarni
- Bioprospecting Group; Agharkar Research Institute; G. G. Agarkar Road Pune 411004 India
| | - Ramasamy Karvembu
- Department of Chemistry; National Institute of Technology; Tiruchirappalli 620015 India
| |
Collapse
|
29
|
Chen GF, Xu TH, Yan Y, Zhou YR, Jiang Y, Melcher K, Xu HE. Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol Sin 2017; 38:1205-1235. [PMID: 28713158 PMCID: PMC5589967 DOI: 10.1038/aps.2017.28] [Citation(s) in RCA: 1186] [Impact Index Per Article: 148.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/02/2017] [Indexed: 12/12/2022]
Abstract
Amyloid beta peptide (Aβ) is produced through the proteolytic processing of a transmembrane protein, amyloid precursor protein (APP), by β- and γ-secretases. Aβ accumulation in the brain is proposed to be an early toxic event in the pathogenesis of Alzheimer's disease, which is the most common form of dementia associated with plaques and tangles in the brain. Currently, it is unclear what the physiological and pathological forms of Aβ are and by what mechanism Aβ causes dementia. Moreover, there are no efficient drugs to stop or reverse the progression of Alzheimer's disease. In this paper, we review the structures, biological functions, and neurotoxicity role of Aβ. We also discuss the potential receptors that interact with Aβ and mediate Aβ intake, clearance, and metabolism. Additionally, we summarize the therapeutic developments and recent advances of different strategies for treating Alzheimer's disease. Finally, we will report on the progress in searching for novel, potentially effective agents as well as selected promising strategies for the treatment of Alzheimer's disease. These prospects include agents acting on Aβ, its receptors and tau protein, such as small molecules, vaccines and antibodies against Aβ; inhibitors or modulators of β- and γ-secretase; Aβ-degrading proteases; tau protein inhibitors and vaccines; amyloid dyes and microRNAs.
Collapse
Affiliation(s)
- Guo-Fang Chen
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ting-Hai Xu
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yan Yan
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yu-Ren Zhou
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yi Jiang
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Karsten Melcher
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - H Eric Xu
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
30
|
Cremades N, Dobson CM. The contribution of biophysical and structural studies of protein self-assembly to the design of therapeutic strategies for amyloid diseases. Neurobiol Dis 2017; 109:178-190. [PMID: 28709995 DOI: 10.1016/j.nbd.2017.07.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/26/2017] [Accepted: 07/10/2017] [Indexed: 01/26/2023] Open
Abstract
Many neurodegenerative disorders, including Alzheimer's, Parkinson's and the prion diseases, are characterized by a conformational conversion of normally soluble proteins or peptides into pathological species, by a process of misfolding and self-assembly that leads ultimately to the formation of amyloid fibrils. Recent studies support the idea that multiple intermediate species with a wide variety of degrees of neuronal toxicity are generated during such processes. The development of a high level of knowledge of the nature and structure of the pathogenic amyloid species would significantly enhance efforts to underline the molecular origins of these disorders and also to develop both accurate diagnoses and effective therapeutic interventions for these types of conditions. In this review, we discuss recent biophysical and structural information concerning different types of amyloid aggregates and the way in which such information can guide rational therapeutic approaches designed to target specific pathogenic events that occur during the development of these highly debilitating and increasingly common diseases.
Collapse
Affiliation(s)
- Nunilo Cremades
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Unit BIFI-IQFR(CSIC), Universidad de Zaragoza, Zaragoza 50018, Spain.
| | - Christopher M Dobson
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
31
|
Asadbegi M, Yaghmaei P, Salehi I, Komaki A, Ebrahim-Habibi A. Investigation of thymol effect on learning and memory impairment induced by intrahippocampal injection of amyloid beta peptide in high fat diet- fed rats. Metab Brain Dis 2017; 32:827-839. [PMID: 28255862 DOI: 10.1007/s11011-017-9960-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/24/2017] [Indexed: 01/28/2023]
Abstract
Obesity and consumption of a high fat diet (HFD) are known to increase the risk of Alzheimer's disease (AD). In the present study, we have examined the protective and therapeutic effects of thymol (main monoterpene phenol found in thyme essential oil) on a HFD-fed rat model of AD. Fourty adult male Wistar rats were randomly assigned to 5 groups:(n = 8 rats/group): group 1, control, consumed an ordinary diet, group 2 consumed a HFD for 8 weeks, then received phosphate-buffered saline (PBS) via intrahippocampal (IHP) injection, group 3 consumed HFD for 8 weeks, then received beta-amyloid (Aβ)1-42 via IHP injections to induce AD, group 4 consumed HFD for 8 weeks, then received Aβ1-42, and was treated by thymol (30 mg/kg in sunflower oil) daily for 4 weeks, and group 5 consumed HFD for 8 week, then received Aβ1-42 after what sunflower oil was administered by oral gavage daily for 4 weeks. Biochemical tests showed an impaired lipid profile and higher glucose levels upon consumption of HFD, which was ameliorated by thymol treatment. In behavioral results, spatial memory in group 3 was significantly impaired, but groups treated with thymol showed better spatial memory compared to group 3 (p ≤ 0.01). In histological results, formation of Aβ plaque in hippocampus of group 3 increased significantly compared to group 1 and group 2 (p ≤ 0.05), but group 4 showed decreased Aβ plaques compared to group 3 (p ≤ 0.01). In conclusion, thymol decreased the effects of Aβ on memory and could be considered as neuroprotective.
Collapse
Affiliation(s)
- Masoumeh Asadbegi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Azadeh Ebrahim-Habibi
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411413137, Iran.
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Seixas da Silva GS, Melo HM, Lourenco MV, Lyra E Silva NM, de Carvalho MB, Alves-Leon SV, de Souza JM, Klein WL, da-Silva WS, Ferreira ST, De Felice FG. Amyloid-β oligomers transiently inhibit AMP-activated kinase and cause metabolic defects in hippocampal neurons. J Biol Chem 2017; 292:7395-7406. [PMID: 28302722 DOI: 10.1074/jbc.m116.753525] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 03/15/2017] [Indexed: 11/06/2022] Open
Abstract
AMP-activated kinase (AMPK) is a key player in energy sensing and metabolic reprogramming under cellular energy restriction. Several studies have linked impaired AMPK function to peripheral metabolic diseases such as diabetes. However, the impact of neurological disorders, such as Alzheimer disease (AD), on AMPK function and downstream effects of altered AMPK activity on neuronal metabolism have been investigated only recently. Here, we report the impact of Aβ oligomers (AβOs), synaptotoxins that accumulate in AD brains, on neuronal AMPK activity. Short-term exposure of cultured rat hippocampal neurons or ex vivo human cortical slices to AβOs transiently decreased intracellular ATP levels and AMPK activity, as evaluated by its phosphorylation at threonine residue 172 (AMPK-Thr(P)172). The AβO-dependent reduction in AMPK-Thr(P)172 levels was mediated by glutamate receptors of the N-methyl-d-aspartate (NMDA) subtype and resulted in removal of glucose transporters (GLUTs) from the surfaces of dendritic processes in hippocampal neurons. Importantly, insulin prevented the AβO-induced inhibition of AMPK. Our results establish a novel toxic impact of AβOs on neuronal metabolism and suggest that AβO-induced, NMDA receptor-mediated AMPK inhibition may play a key role in early brain metabolic defects in AD.
Collapse
Affiliation(s)
| | - Helen M Melo
- From the Institute of Medical Biochemistry Leopoldo de Meis and
| | - Mychael V Lourenco
- From the Institute of Medical Biochemistry Leopoldo de Meis and.,the Institute of Biophysics Carlos Chagas Filho
| | | | | | | | - Jorge M de Souza
- Neurosurgery, Clementino Fraga Filho Hospital, Federal University of Rio De Janeiro, Rio de Janeiro 21941-902, Brazil
| | - William L Klein
- the Department of Neurobiology, Northwestern University, Evanston, Illinois 60208-3520, and
| | | | - Sergio T Ferreira
- From the Institute of Medical Biochemistry Leopoldo de Meis and.,the Institute of Biophysics Carlos Chagas Filho
| | - Fernanda G De Felice
- From the Institute of Medical Biochemistry Leopoldo de Meis and .,the Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
33
|
Wu X, Kosaraju J, Zhou W, Tam KY. Neuroprotective Effect of SLM, a Novel Carbazole-Based Fluorophore, on SH-SY5Y Cell Model and 3xTg-AD Mouse Model of Alzheimer's Disease. ACS Chem Neurosci 2017; 8:676-685. [PMID: 28032988 DOI: 10.1021/acschemneuro.6b00388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Amyloid β (Aβ) peptide aggregating to form a neurotoxic plaque, leading to cognitive deficits, is believed to be one of the plausible mechanisms for Alzheimer's disease (AD). Inhibiting Aβ aggregation is supposed to offer a neuroprotective effect to ameliorate AD. A previous report has shown that SLM, a carbazole-based fluorophore, binds to Aβ to inhibit the aggregation. However, it is not entirely clear whether the inhibition of Aβ aggregation alone would lead to the anticipated neuroprotective effects. In the current study, we intended to examine the protective action of SLM against Aβ-induced neurotoxicity in vitro and to evaluate if SLM can decrease the cognitive and behavioral deficits observed in triple transgenic AD mouse model (3xTg-AD). In the in vitro study, neurotoxicity induced by Aβ42 in human neuroblastoma (SH-SY5Y) cells was found to be reduced through the treatment with SLM. In the in vivo study, following one month SLM intraperitoneal injection (1, 2, and 4 mg/kg), 3xTg-AD mice were tested on Morris water maze (MWM) and Y-maze for their cognitive ability and sacrificed for biochemical estimations. Results show that SLM treatment improved the learning and memory ability in 3xTg-AD mice in MWM and Y-maze tasks. SLM also mitigated the amyloid burden by decreasing brain Aβ40 and Aβ42 levels and reduced tau phosphorylation, glycogen synthase kinase-3β activity, and neuro-inflammation. From our observations, SLM shows neuroprotection in SH-SY5Y cells against Aβ42 and also in 3xTg-AD mouse model by mitigating the pathological features and behavioral impairments.
Collapse
Affiliation(s)
- Xiaoli Wu
- Drug Development Core, Faculty
of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jayasankar Kosaraju
- Drug Development Core, Faculty
of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wei Zhou
- Drug Development Core, Faculty
of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kin Yip Tam
- Drug Development Core, Faculty
of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
34
|
Giacomelli C, Daniele S, Martini C. Potential biomarkers and novel pharmacological targets in protein aggregation-related neurodegenerative diseases. Biochem Pharmacol 2017; 131:1-15. [PMID: 28159621 DOI: 10.1016/j.bcp.2017.01.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
The aggregation of specific proteins plays a pivotal role in the etiopathogenesis of several neurodegenerative diseases (NDs). β-Amyloid (Aβ) peptide-containing plaques and intraneuronal neurofibrillary tangles composed of hyperphosphorylated protein tau are the two main neuropathological lesions in Alzheimer's disease. Meanwhile, Parkinson's disease is defined by the presence of intraneuronal inclusions (Lewy bodies), in which α-synuclein (α-syn) has been identified as a major protein component. The current literature provides considerable insights into the mechanisms underlying oligomeric-related neurodegeneration, as well as the relationship between protein aggregation and ND, thus facilitating the development of novel putative biomarkers and/or pharmacological targets. Recently, α-syn, tau and Aβ have been shown to interact each other or with other "pathological proteins" to form toxic heteroaggregates. These latest findings are overcoming the concept that each neurodegenerative disease is related to the misfolding of a single specific protein. In this review, potential opportunities and pharmacological approaches targeting α-syn, tau and Aβ and their oligomeric forms are highlighted with examples from recent studies. Protein aggregation as a biomarker of NDs, in both the brain and peripheral fluids, is deeply explored. Finally, the relationship between biomarker establishment and assessment and their use as diagnostics or therapeutic targets are discussed.
Collapse
Affiliation(s)
- Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy.
| |
Collapse
|
35
|
Basu A, Suresh Kumar G. Interaction and inhibitory influence of the azo dye carmoisine on lysozyme amyloid fibrillogenesis. MOLECULAR BIOSYSTEMS 2017. [DOI: 10.1039/c7mb00207f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The azo dye carmoisine has a significant inhibitory effect on fibrillogenesis in lysozyme.
Collapse
Affiliation(s)
- Anirban Basu
- Biophysical Chemistry Laboratory
- Organic & Medicinal Chemistry Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata 700 032
- India
| | - Gopinatha Suresh Kumar
- Biophysical Chemistry Laboratory
- Organic & Medicinal Chemistry Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata 700 032
- India
| |
Collapse
|
36
|
A comparative study of fibrillation kinetics of two homologous proteins under identical solution condition. Biochimie 2017; 132:75-84. [DOI: 10.1016/j.biochi.2016.11.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/03/2016] [Indexed: 02/08/2023]
|
37
|
Ramshini H, Mannini B, Khodayari K, Ebrahim-Habibi A, Moghaddasi AS, Tayebee R, Chiti F. Bis(indolyl)phenylmethane derivatives are effective small molecules for inhibition of amyloid fibril formation by hen lysozyme. Eur J Med Chem 2016; 124:361-371. [DOI: 10.1016/j.ejmech.2016.08.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 12/31/2022]
|
38
|
Mao Y, Guo Z, Zheng T, Jiang Y, Yan Y, Yin X, Chen Y, Zhang B. Intranasal insulin alleviates cognitive deficits and amyloid pathology in young adult APPswe/PS1dE9 mice. Aging Cell 2016; 15:893-902. [PMID: 27457264 PMCID: PMC5013027 DOI: 10.1111/acel.12498] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2016] [Indexed: 11/26/2022] Open
Abstract
Brain insulin signaling deficits contribute to multiple pathological features of Alzheimer's disease (AD). Although intranasal insulin has shown efficacy in patients with AD, the underlying mechanisms remain largely unillustrated. Here, we demonstrate that intranasal insulin improves cognitive deficits, ameliorates defective brain insulin signaling, and strongly reduces β‐amyloid (Aβ) production and plaque formation after 6 weeks of treatment in 4.5‐month‐old APPswe/PS1dE9 (APP/PS1) mice. Furthermore, c‐Jun N‐terminal kinase activation, which plays a pivotal role in insulin resistance and AD pathologies, is significantly inhibited. The alleviation of amyloid pathology by intranasal insulin results mainly from enhanced nonamyloidogenic processing and compromised amyloidogenic processing of amyloid precursor protein (APP), and from a reduction in apolipoprotein E protein which is involved in Aβ metabolism. In addition, intranasal insulin effectively promotes hippocampal neurogenesis in APP/PS1 mice. This study, exploring the mechanisms underlying the beneficial effects of intranasal insulin on Aβ pathologies in vivo for the first time, highlights important preclinical evidence that intranasal insulin is potentially an effective therapeutic method for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yan‐Fang Mao
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Zhangyu Guo
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Tingting Zheng
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yasi Jiang
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yaping Yan
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Xinzhen Yin
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yanxing Chen
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Baorong Zhang
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| |
Collapse
|
39
|
Effect of curcumin analogs onα-synuclein aggregation and cytotoxicity. Sci Rep 2016; 6:28511. [PMID: 27338805 PMCID: PMC4919791 DOI: 10.1038/srep28511] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/06/2016] [Indexed: 01/17/2023] Open
Abstract
Alpha-synuclein (α-Syn) aggregation into oligomers and fibrils is associated with dopaminergic neuron loss occurring in Parkinson’s disease (PD) pathogenesis. Compounds that modulate α-Syn aggregation and interact with preformed fibrils/oligomers and convert them to less toxic species could have promising applications in the drug development efforts against PD. Curcumin is one of the Asian food ingredient which showed promising role as therapeutic agent against many neurological disorders including PD. However, the instability and low solubility makes it less attractive for the drug development. In this work, we selected various curcumin analogs and studied their toxicity, stability and efficacy to interact with different α-Syn species and modulation of their toxicity. We found a subset of curcumin analogs with higher stability and showed that curcumin and its various analogs interact with preformed fibrils and oligomers and accelerate α-Syn aggregation to produce morphologically different amyloid fibrils in vitro. Furthermore, these curcumin analogs showed differential binding with the preformed α-Syn aggregates. The present data suggest the potential role of curcumin analogs in modulating α-Syn aggregation.
Collapse
|
40
|
Structure of amyloid oligomers and their mechanisms of toxicities: Targeting amyloid oligomers using novel therapeutic approaches. Eur J Med Chem 2016; 114:41-58. [DOI: 10.1016/j.ejmech.2016.02.065] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 01/22/2023]
|
41
|
Sengupta U, Nilson AN, Kayed R. The Role of Amyloid-β Oligomers in Toxicity, Propagation, and Immunotherapy. EBioMedicine 2016; 6:42-49. [PMID: 27211547 PMCID: PMC4856795 DOI: 10.1016/j.ebiom.2016.03.035] [Citation(s) in RCA: 547] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/03/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023] Open
Abstract
The incidence of Alzheimer's disease (AD) is growing every day and finding an effective treatment is becoming more vital. Amyloid-β (Aβ) has been the focus of research for several decades. The recent shift in the Aβ cascade hypothesis from all Aβ to small soluble oligomeric intermediates is directing the search for therapeutics towards the toxic mediators of the disease. Targeting the most toxic oligomers may prove to be an effective treatment by preventing their spread. Specific targeting of oligomers has been shown to protect cognition in rodent models. Additionally, the heterogeneity of research on Aβ oligomers may seem contradictory until size and conformation are taken into account. In this review, we will discuss Aβ oligomers and their toxicity in relation to size and conformation as well as their influence on inflammation and the potential of Aβ oligomer immunotherapy.
Collapse
Affiliation(s)
- Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ashley N Nilson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
42
|
Zheng Y, Tian S, Peng X, Yang J, Fu Y, Jiao Y, Zhao J, He J, Hong T. Kinesin-1 inhibits the aggregation of amyloid-β peptide as detected by fluorescence cross-correlation spectroscopy. FEBS Lett 2016; 590:1028-37. [DOI: 10.1002/1873-3468.12137] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 02/04/2016] [Accepted: 03/11/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Yanpeng Zheng
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; China
| | - Shijun Tian
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; China
| | - Xianglei Peng
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; China
| | - Jingfa Yang
- Beijing National Laboratory for Molecular Science; Institute of Chemistry; Chinese Academy of Sciences; Beijing China
| | - Yuanhui Fu
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; China
| | - Yueying Jiao
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; China
| | - Jiang Zhao
- Beijing National Laboratory for Molecular Science; Institute of Chemistry; Chinese Academy of Sciences; Beijing China
| | - Jinsheng He
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; China
| | - Tao Hong
- College of Life Sciences and Bioengineering; Beijing Jiaotong University; China
- Institute for Viral Disease Control and Prevention; Chinese Centre for Disease Control and Prevention; Beijing China
| |
Collapse
|
43
|
Young LM, Saunders JC, Mahood RA, Revill CH, Foster RJ, Ashcroft AE, Radford SE. ESI-IMS-MS: A method for rapid analysis of protein aggregation and its inhibition by small molecules. Methods 2016; 95:62-9. [PMID: 26007606 PMCID: PMC4769093 DOI: 10.1016/j.ymeth.2015.05.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/26/2015] [Accepted: 05/07/2015] [Indexed: 11/21/2022] Open
Abstract
Electrospray ionisation-ion mobility spectrometry-mass spectrometry (ESI-IMS-MS) is a powerful method for the study of conformational changes in protein complexes, including oligomeric species populated during protein self-aggregation into amyloid fibrils. Information on the mass, stability, cross-sectional area and ligand binding capability of each transiently populated intermediate, present in the heterogeneous mixture of assembling species, can be determined individually in a single experiment in real-time. Determining the structural characterisation of oligomeric species and alterations in self-assembly pathways observed in the presence of small molecule inhibitors is of great importance, given the urgent demand for effective therapeutics. Recent studies have demonstrated the capability of ESI-IMS-MS to identify small molecule modulators of amyloid assembly and to determine the mechanism by which they interact (positive, negative, non-specific binding, or colloidal) in a high-throughput format. Here, we demonstrate these advances using self-assembly of Aβ40 as an example, and reveal two new inhibitors of Aβ40 fibrillation.
Collapse
Affiliation(s)
- Lydia M Young
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, United Kingdom.
| | - Janet C Saunders
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, United Kingdom.
| | - Rachel A Mahood
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, United Kingdom.
| | - Charlotte H Revill
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; School of Chemistry, University of Leeds, LS2 9JT, United Kingdom.
| | - Richard J Foster
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; School of Chemistry, University of Leeds, LS2 9JT, United Kingdom.
| | - Alison E Ashcroft
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, United Kingdom.
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, United Kingdom.
| |
Collapse
|
44
|
How SC, Yang SM, Hsin A, Tseng CP, Hsueh SS, Lin MS, Chen RPY, Chou WL, Wang SSS. Examining the inhibitory potency of food additive fast green FCF against amyloid fibrillogenesis under acidic conditions. Food Funct 2016; 7:4898-4907. [DOI: 10.1039/c6fo00792a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Amyloid fibril formation of hen lysozyme (HEWL) can be attenuated by fast green FCF.
Collapse
Affiliation(s)
- Su-Chun How
- Department of Chemical Engineering
- National Taiwan University
- Taipei 10617
- Taiwan
| | - Szu-Ming Yang
- Department of Chemical Engineering
- National Taiwan University
- Taipei 10617
- Taiwan
| | - Ai Hsin
- Department of Chemical Engineering
- National Taiwan University
- Taipei 10617
- Taiwan
| | - Chia-Ping Tseng
- Department of Chemical Engineering
- National Taiwan University
- Taipei 10617
- Taiwan
| | - Shu-Shun Hsueh
- Department of Chemical Engineering
- National Taiwan University
- Taipei 10617
- Taiwan
| | | | - Rita P.-Y. Chen
- Institute of Biochemical Sciences
- National Taiwan University
- Taipei 10617
- Taiwan
- Institute of Biological Chemistry
| | - Wei-Lung Chou
- Department of Safety
- Health and Environmental Engineering
- Hungkuang University
- Taichung City 433
- Taiwan
| | - Steven S.-S. Wang
- Department of Chemical Engineering
- National Taiwan University
- Taipei 10617
- Taiwan
| |
Collapse
|
45
|
Shariatizi S, Meratan AA, Ghasemi A, Nemat-Gorgani M. Inhibition of amyloid fibrillation and cytotoxicity of lysozyme fibrillation products by polyphenols. Int J Biol Macromol 2015; 80:95-106. [PMID: 26102331 DOI: 10.1016/j.ijbiomac.2015.06.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/13/2022]
Abstract
An increasing number of studies conducted under in vitro and in vivo conditions, have concluded that polyphenols, compounds frequently occurring in many herbs with antioxidant properties, prevent and reverse amyloid fibril formation. However, the mechanisms by which these natural products modulate the protein aggregation process are poorly understood. Herein, a range of techniques including thioflavin T (ThT) and ANS fluorescence assays, electron microscopy and circular dichroism have been employed to determine the efficacy of rosmarinic acid (RA) and resveratrol (Res) on the inhibition/reversion of fibrillogenesis and hindering cytotoxicity induced by protofibrils and amyloid fibrils of hen egg white lysozyme (HEWL). Results demonstrated that both polyphenols effectively inhibit fibrillogenesis and destabilize preformed fibrils of HEWL in a concentration-dependent manner. Cytotoxicity protection on PC12 cells was also observed using the MTT assay, ROS production assay, and phase-contrast microscopy. It is suggested that the mechanism underlying the inhibitory effects of RA and Res is to prevent hydrophobic interactions between HEWL amyloidogenic prefibrillar species, although additional studies is needed to elucidate the detailed mechanisms involved. A combination of antioxidative and anti-amyloidogenic properties of these molecules may provide them with the described neuroprotective capacities.
Collapse
Affiliation(s)
- Sajad Shariatizi
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, 1417614411 Tehran, Iran
| | - Ali Akbar Meratan
- Department of Biotechnology, Ramin University of Agricultural and Natural Resources, Khouzestan, Iran.
| | - Atiyeh Ghasemi
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, 1417614411 Tehran, Iran
| | | |
Collapse
|
46
|
Pachahara SK, Adicherla H, Nagaraj R. Self-Assembly of Aβ40, Aβ42 and Aβ43 Peptides in Aqueous Mixtures of Fluorinated Alcohols. PLoS One 2015; 10:e0136567. [PMID: 26308214 PMCID: PMC4550328 DOI: 10.1371/journal.pone.0136567] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 08/04/2015] [Indexed: 12/02/2022] Open
Abstract
Fluorinated alcohols such as hexafluoroisopropanol (HFIP) and trifluoroethanol (TFE) have the ability to promote α-helix and β-hairpin structure in proteins and peptides. HFIP has been used extensively to dissolve various amyloidogenic proteins and peptides including Aβ, in order to ensure their monomeric status. In this paper, we have investigated the self-assembly of Aβ40, Aβ42, and Aβ43 in aqueous mixtures of fluorinated alcohols from freshly dissolved stock solutions in HFIP. We have observed that formation of fibrillar and non-fibrillar structures are dependent on the solvent composition. Peptides form fibrils with ease when reconstituted in deionized water from freshly dissolved HFIP stocks. In aqueous mixtures of fluorinated alcohols, either predominant fibrillar structures or clustered aggregates were observed. Aqueous mixtures of 20% HFIP are more favourable for Aβ fibril formation as compared to 20% TFE. When Aβ40, Aβ42, and Aβ43 stocks in HFIP are diluted in 50% aqueous mixtures in phosphate buffer or deionized water followed by slow evaporation of HFIP, Aβ peptides form fibrils in phosphate buffer and deionized water. The clustered structures could be off-pathway aggregates. Aβ40, Aβ42, and Aβ43 showed significant α-helical content in freshly dissolved HFIP stocks. The α-helical conformational intermediate in Aβ40, Aβ42, and Aβ43 could favour the formation of both fibrillar and non-fibrillar aggregates depending on solvent conditions and rate of α-helical to β-sheet transition.
Collapse
Affiliation(s)
| | - Harikrishna Adicherla
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500 007, India
| | - Ramakrishnan Nagaraj
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500 007, India
- * E-mail:
| |
Collapse
|
47
|
Ferreira ST, Lourenco MV, Oliveira MM, De Felice FG. Soluble amyloid-β oligomers as synaptotoxins leading to cognitive impairment in Alzheimer's disease. Front Cell Neurosci 2015; 9:191. [PMID: 26074767 PMCID: PMC4443025 DOI: 10.3389/fncel.2015.00191] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/30/2015] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia in the elderly, and affects millions of people worldwide. As the number of AD cases continues to increase in both developed and developing countries, finding therapies that effectively halt or reverse disease progression constitutes a major research and public health challenge. Since the identification of the amyloid-β peptide (Aβ) as the major component of the amyloid plaques that are characteristically found in AD brains, a major effort has aimed to determine whether and how Aβ leads to memory loss and cognitive impairment. A large body of evidence accumulated in the past 15 years supports a pivotal role of soluble Aβ oligomers (AβOs) in synapse failure and neuronal dysfunction in AD. Nonetheless, a number of basic questions, including the exact molecular composition of the synaptotoxic oligomers, the identity of the receptor(s) to which they bind, and the signaling pathways that ultimately lead to synapse failure, remain to be definitively answered. Here, we discuss recent advances that have illuminated our understanding of the chemical nature of the toxic species and the deleterious impact they have on synapses, and have culminated in the proposal of an Aβ oligomer hypothesis for Alzheimer’s pathogenesis. We also highlight outstanding questions and challenges in AD research that should be addressed to allow translation of research findings into effective AD therapies.
Collapse
Affiliation(s)
- Sergio T Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil ; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Mauricio M Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| |
Collapse
|
48
|
Porzoor A, Alford B, Hügel HM, Grando D, Caine J, Macreadie I. Anti-amyloidogenic properties of some phenolic compounds. Biomolecules 2015; 5:505-27. [PMID: 25898401 PMCID: PMC4496683 DOI: 10.3390/biom5020505] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/02/2015] [Accepted: 04/03/2015] [Indexed: 12/23/2022] Open
Abstract
A family of 21 polyphenolic compounds consisting of those found naturally in danshen and their analogues were synthesized and subsequently screened for their anti-amyloidogenic activity against the amyloid beta peptide (Aβ42) of Alzheimer’s disease. After 24 h incubation with Aβ42, five compounds reduced thioflavin T (ThT) fluorescence, indicative of their anti-amyloidogenic propensity (p < 0.001). TEM and immunoblotting analysis also showed that selected compounds were capable of hindering fibril formation even after prolonged incubations. These compounds were also capable of rescuing the yeast cells from toxic changes induced by the chemically synthesized Aβ42. In a second assay, a Saccharomyces cerevisiae AHP1 deletant strain transformed with GFP fused to Aβ42 was treated with these compounds and analyzed by flow cytometry. There was a significant reduction in the green fluorescence intensity associated with 14 compounds. We interpret this result to mean that the compounds had an anti-amyloid-aggregation propensity in the yeast and GFP-Aβ42 was removed by proteolysis. The position and not the number of hydroxyl groups on the aromatic ring was found to be the most important determinant for the anti-amyloidogenic properties.
Collapse
Affiliation(s)
- Afsaneh Porzoor
- School of Applied Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Benjamin Alford
- School of Applied Sciences, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Helmut M Hügel
- School of Applied Sciences, RMIT University, Melbourne, Victoria 3000, Australia.
| | - Danilla Grando
- School of Applied Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Joanne Caine
- Materials Science and Engineering, CSIRO Preventative Health Flagship, 343 Royal Parade, Parkville, Victoria 3052, Australia.
| | - Ian Macreadie
- School of Applied Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
49
|
Singh PK, Ghosh D, Tewari D, Mohite GM, Carvalho E, Jha NN, Jacob RS, Sahay S, Banerjee R, Bera AK, Maji SK. Cytotoxic helix-rich oligomer formation by melittin and pancreatic polypeptide. PLoS One 2015; 10:e0120346. [PMID: 25803428 PMCID: PMC4372375 DOI: 10.1371/journal.pone.0120346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/21/2015] [Indexed: 12/20/2022] Open
Abstract
Conversion of amyloid fibrils by many peptides/proteins involves cytotoxic helix-rich oligomers. However, their toxicity and biophysical studies remain largely unknown due to their highly dynamic nature. To address this, we chose two helical peptides (melittin, Mel and pancreatic polypeptide, PP) and studied their aggregation and toxicity. Mel converted its random coil structure to oligomeric helical structure upon binding to heparin; however, PP remained as helix after oligomerization. Interestingly, similar to Parkinson’s associated α-synuclein (AS) oligomers, Mel and PP also showed tinctorial properties, higher hydrophobic surface exposure, cellular toxicity and membrane pore formation after oligomerization in the presence of heparin. We suggest that helix-rich oligomers with exposed hydrophobic surface are highly cytotoxic to cells irrespective of their disease association. Moreover as Mel and PP (in the presence of heparin) instantly self-assemble into stable helix-rich amyloidogenic oligomers; they could be represented as models for understanding the biophysical and cytotoxic properties of helix-rich intermediates in detail.
Collapse
Affiliation(s)
- Pradeep K. Singh
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Dhiman Ghosh
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Debanjan Tewari
- Department of Biotechnology, IIT Madras, Chennai, Tamil Nadu, India
| | - Ganesh M. Mohite
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Edmund Carvalho
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Narendra Nath Jha
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Reeba S. Jacob
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Shruti Sahay
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Amal K. Bera
- Department of Biotechnology, IIT Madras, Chennai, Tamil Nadu, India
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
- * E-mail:
| |
Collapse
|
50
|
Young LM, Saunders JC, Mahood RA, Revill CH, Foster RJ, Tu LH, Raleigh DP, Radford SE, Ashcroft AE. Screening and classifying small-molecule inhibitors of amyloid formation using ion mobility spectrometry-mass spectrometry. Nat Chem 2014; 7:73-81. [PMID: 25515893 PMCID: PMC4280571 DOI: 10.1038/nchem.2129] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/05/2014] [Indexed: 12/28/2022]
Abstract
The search for therapeutic agents which bind specifically to precursor protein conformations and inhibit amyloid assembly is an important challenge. Identifying such inhibitors is difficult since many protein precursors of aggregation are partially folded or intrinsically disordered, ruling out structure-based design. Furthermore, inhibitors can act by a variety of mechanisms, including specific or non-specific binding, as well as colloidal inhibition. Here we report a high throughput method based on ion mobility spectrometry-mass spectrometry (IMS-MS) that is capable of rapidly detecting small molecules that bind to amyloid precursors, identifying the interacting protein species, and defining the mode of inhibition. Using this method we have classified a variety of small molecules that are potential inhibitors of human islet amyloid polypeptide (hIAPP) aggregation or amyloid-beta 1-40 (Aβ40) aggregation as either specific, non-specific, colloidal or non-interacting. We also demonstrate the ability of IMS-MS to screen for inhibitory small molecules in a 96-well plate format and use this to discover a new inhibitor of hIAPP amyloid assembly.
Collapse
Affiliation(s)
- Lydia M Young
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK.,School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK
| | - Janet C Saunders
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK.,School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK
| | - Rachel A Mahood
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK.,School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK
| | - Charlotte H Revill
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK.,School of Chemistry, University of Leeds, LS2 9JT, UK
| | - Richard J Foster
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK.,School of Chemistry, University of Leeds, LS2 9JT, UK
| | - Ling-Hsien Tu
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Daniel P Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK.,School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK
| | - Alison E Ashcroft
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK.,School of Molecular and Cellular Biology, University of Leeds, LS2 9JT, UK
| |
Collapse
|