51
|
Eccleston RC, Pollock DD, Goldstein RA. Selection for cooperativity causes epistasis predominately between native contacts and enables epistasis-based structure reconstruction. Proc Natl Acad Sci U S A 2021; 118:e2010057118. [PMID: 33879570 PMCID: PMC8072402 DOI: 10.1073/pnas.2010057118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epistasis and cooperativity of folding both result from networks of energetic interactions in proteins. Epistasis results from energetic interactions among mutants, whereas cooperativity results from energetic interactions during folding that reduce the presence of intermediate states. The two concepts seem intuitively related, but it is unknown how they are related, particularly in terms of selection. To investigate their relationship, we simulated protein evolution under selection for cooperativity and separately under selection for epistasis. Strong selection for cooperativity created strong epistasis between contacts in the native structure but weakened epistasis between nonnative contacts. In contrast, selection for epistasis increased epistasis in both native and nonnative contacts and reduced cooperativity. Because epistasis can be used to predict protein structure only if it preferentially occurs in native contacts, this result indicates that selection for cooperativity may be key for predicting structure using epistasis. To evaluate this inference, we simulated the evolution of guanine nucleotide-binding protein (GB1) with and without cooperativity. With cooperativity, strong epistatic interactions clearly map out the native GB1 structure, while allowing the presence of intermediate states (low cooperativity) obscured the structure. This indicates that using epistasis measurements to reconstruct protein structure may be inappropriate for proteins with stable intermediates.
Collapse
Affiliation(s)
- R Charlotte Eccleston
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - David D Pollock
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Richard A Goldstein
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom;
| |
Collapse
|
52
|
Oyarzún MP, Tapia-Arellano A, Cabrera P, Jara-Guajardo P, Kogan MJ. Plasmonic Nanoparticles as Optical Sensing Probes for the Detection of Alzheimer's Disease. SENSORS (BASEL, SWITZERLAND) 2021; 21:2067. [PMID: 33809416 PMCID: PMC7998661 DOI: 10.3390/s21062067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), considered a common type of dementia, is mainly characterized by a progressive loss of memory and cognitive functions. Although its cause is multifactorial, it has been associated with the accumulation of toxic aggregates of the amyloid-β peptide (Aβ) and neurofibrillary tangles (NFTs) of tau protein. At present, the development of highly sensitive, high cost-effective, and non-invasive diagnostic tools for AD remains a challenge. In the last decades, nanomaterials have emerged as an interesting and useful tool in nanomedicine for diagnostics and therapy. In particular, plasmonic nanoparticles are well-known to display unique optical properties derived from their localized surface plasmon resonance (LSPR), allowing their use as transducers in various sensing configurations and enhancing detection sensitivity. Herein, this review focuses on current advances in in vitro sensing techniques such as Surface-enhanced Raman scattering (SERS), Surface-enhanced fluorescence (SEF), colorimetric, and LSPR using plasmonic nanoparticles for improving the sensitivity in the detection of main biomarkers related to AD in body fluids. Additionally, we refer to the use of plasmonic nanoparticles for in vivo imaging studies in AD.
Collapse
Affiliation(s)
- María Paz Oyarzún
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Andreas Tapia-Arellano
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Pablo Cabrera
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Pedro Jara-Guajardo
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Marcelo J. Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| |
Collapse
|
53
|
Malishev R, Ben-Zichri S, Oren O, Shauloff N, Peretz T, Taube R, Papo N, Jelinek R. The pro-apoptotic domain of BIM protein forms toxic amyloid fibrils. Cell Mol Life Sci 2021; 78:2145-2155. [PMID: 32844279 PMCID: PMC11072030 DOI: 10.1007/s00018-020-03623-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/26/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022]
Abstract
BIM is a key apoptotic protein, participating in diverse cellular processes. Interestingly, recent studies have hypothesized that BIM is associated with the extensive neuronal cell death encountered in protein misfolding diseases, such as Alzheimer's disease. Here, we report that the core pro-apoptotic domain of BIM, the BIM-BH3 motif, forms ubiquitous amyloid fibrils. The BIM-BH3 fibrils exhibit cytotoxicity, disrupt mitochondrial functions, and modulate the structures and dynamics of mitochondrial membrane mimics. Interestingly, a slightly longer peptide in which BIM-BH3 was flanked by four additional residues, widely employed as a model of the pro-apoptotic core domain of BIM, did not form fibrils, nor exhibited cell disruptive properties. The experimental data suggest a new mechanistic role for the BIM-BH3 domain, and demonstrate, for the first time, that an apoptotic peptide forms toxic amyloid fibrils.
Collapse
Affiliation(s)
- Ravit Malishev
- Department of Chemistry and Ilse, Katz Institute for Nanotechnology, Ben Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Shani Ben-Zichri
- Department of Chemistry and Ilse, Katz Institute for Nanotechnology, Ben Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Ofek Oren
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Nitzan Shauloff
- Department of Chemistry and Ilse, Katz Institute for Nanotechnology, Ben Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Tal Peretz
- Department of Life Sciences, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Ran Taube
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Raz Jelinek
- Department of Chemistry and Ilse, Katz Institute for Nanotechnology, Ben Gurion University of the Negev, 84105, Beer Sheva, Israel.
| |
Collapse
|
54
|
The concept of protein folding/unfolding and its impacts on human health. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021. [PMID: 34090616 DOI: 10.1016/bs.apcsb.2021.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Proteins have evolved in specific 3D structures and play different functions in cells and determine various reactions and pathways. The newly synthesized amino acid chains once depart ribosome must crumple into three-dimensional structures so can be biologically active. This process of protein that makes a functional molecule is called protein folding. The protein folding is both a biological and a physicochemical process that depends on the sequence of it. In fact, this process occurs more complicated and in some cases and in exposure to some molecules like glucose (glycation), mistaken folding leads to amyloid structures and fatal disorders called conformational diseases. Such conditions are detected by the quality control system of the cell and these abnormal proteins undergo renovation or degradation. This scenario takes place by the chaperones, chaperonins, and Ubiquitin-proteasome complex. Understanding of protein folding mechanisms from different views including experimental and computational approaches has revealed some intermediate ensembles such as molten globule and has been subjected to biophysical and molecular biology attempts to know more about prevalent conformational diseases.
Collapse
|
55
|
AA amyloid fibrils from diseased tissue are structurally different from in vitro formed SAA fibrils. Nat Commun 2021; 12:1013. [PMID: 33579941 PMCID: PMC7881110 DOI: 10.1038/s41467-021-21129-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/07/2021] [Indexed: 01/08/2023] Open
Abstract
Systemic AA amyloidosis is a world-wide occurring protein misfolding disease of humans and animals. It arises from the formation of amyloid fibrils from serum amyloid A (SAA) protein. Using cryo electron microscopy we here show that amyloid fibrils which were purified from AA amyloidotic mice are structurally different from fibrils formed from recombinant SAA protein in vitro. Ex vivo amyloid fibrils consist of fibril proteins that contain more residues within their ordered parts and possess a higher β-sheet content than in vitro fibril proteins. They are also more resistant to proteolysis than their in vitro formed counterparts. These data suggest that pathogenic amyloid fibrils may originate from proteolytic selection, allowing specific fibril morphologies to proliferate and to cause damage to the surrounding tissue. Systemic AA amyloidosis is a protein misfolding disease caused by the formation of amyloid fibrils from serum amyloid A (SAA) protein. Here, the authors present the cryo-EM structures of AA amyloid fibrils isolated from mouse tissue and in vitro formed fibrils, which differ in their structures and they also show that the ex vivo fibrils are more resistant to proteolysis than the in vitro fibrils and propose that pathogenic amyloid fibrils might originate from proteolytic selection.
Collapse
|
56
|
Grigolato F, Arosio P. The role of surfaces on amyloid formation. Biophys Chem 2021; 270:106533. [PMID: 33529995 DOI: 10.1016/j.bpc.2020.106533] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023]
Abstract
Interfaces can strongly accelerate or inhibit protein aggregation, destabilizing proteins that are stable in solution or, conversely, stabilizing proteins that are aggregation-prone. Although this behaviour is well-known, our understanding of the molecular mechanisms underlying surface-induced protein aggregation is still largely incomplete. A major challenge is represented by the high number of physico-chemical parameters involved, which are highly specific to the considered combination of protein, surface properties, and solution conditions. The key aspect determining the role of interfaces is the relative propensity of the protein to aggregate at the surface with respect to bulk. In this review, we discuss the multiple molecular determinants that regulate this balance. We summarize current experimental techniques aimed at characterizing protein aggregation at interfaces, and highlight the need to complement experimental analysis with theoretical modelling. In particular, we illustrate how chemical kinetic analysis can be combined with experimental methods to provide insights into the molecular mechanisms underlying surface-induced protein aggregation, under both stagnant and agitation conditions. We summarize recent progress in the study of important amyloids systems, focusing on selected relevant interfaces.
Collapse
Affiliation(s)
- Fulvio Grigolato
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich 8093, Switzerland
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich 8093, Switzerland.
| |
Collapse
|
57
|
Kumar A, Balbach J. Inactivation of parathyroid hormone: perspectives of drug discovery to combating hyperparathyroidism. Curr Mol Pharmacol 2021; 15:292-305. [PMID: 33573587 DOI: 10.2174/1874467214666210126112839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 11/22/2022]
Abstract
Hormonal coordination is tightly regulated within the human body and thus regulates human physiology. The parathyroid hormone (PTH), a member of the endocrine system, regulates the calcium and phosphate level within the human body. Under non-physiological conditions, PTH levels get upregulated (hyperparathyroidism) or downregulated (hypoparathyroidism) due to external or internal factors. In the case of hyperparathyroidism, elevated PTH stimulates cellular receptors present in the bones, kidneys, and intestines to increase the blood calcium level, leading to calcium deposition. This eventually causes various symptoms including kidney stones. Currently, there is no known medication that directly targets PTH in order to suppress its function. Therefore, it is of great interest to find novel small molecules or any other means that can modulate PTH function. The molecular signaling of PTH starts by binding of its N-terminus to the G-protein coupled PTH1/2 receptor. Therefore, any intervention that affects the N-terminus of PTH could be a lead candidate for treating hyperparathyroidism. As a proof-of-concept, there are various possibilities to inhibit molecular PTH function by (i) a small molecule, (ii) N-terminal PTH phosphorylation, (iii) fibril formation and (iv) residue-specific mutations. These modifications put PTH into an inactive state, which will be discussed in detail in this review article. We anticipate that exploring small molecules or other means that affect the N-terminus of PTH could be lead candidates in combating hyperparathyroidism.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College of Science, Technology and Medicine London, South Kensington, London SW7 2BU. United Kingdom
| | - Jochen Balbach
- Institute of Physics, Biophysics, Martin-Luther-University Halle- Wittenberg. Germany
| |
Collapse
|
58
|
Liu SC, Ying YL, Li WH, Wan YJ, Long YT. Snapshotting the transient conformations and tracing the multiple pathways of single peptide folding using a solid-state nanopore. Chem Sci 2021; 12:3282-3289. [PMID: 34164097 PMCID: PMC8179386 DOI: 10.1039/d0sc06106a] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A fundamental question relating to protein folding/unfolding is the time evolution of the folding of a protein into its precisely defined native structure. The proper identification of transition conformations is essential for accurately describing the dynamic protein folding/unfolding pathways. Owing to the rapid transitions and sub-nm conformation differences involved, the acquisition of the transient conformations and dynamics of proteins is difficult due to limited instrumental resolution. Using the electrochemical confinement effect of a solid-state nanopore, we were able to snapshot the transient conformations and trace the multiple transition pathways of a single peptide inside a nanopore. By combining the results with a Markov chain model, this new single-molecule technique is applied to clarify the transition pathways of the β-hairpin peptide, which shows nonequilibrium fluctuations among several blockage current stages. This method enables the high-throughput investigation of transition pathways experimentally to access previously obscure peptide dynamics, which is significant for understanding the folding/unfolding mechanisms and misfolding of peptides or proteins. A solid-state nanopore based method is described for resolving protein-folding-related problems via snapshotting the folding intermediates and characterizing the kinetics of a single peptide.![]()
Collapse
Affiliation(s)
- Shao-Chuang Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China .,Department of Chemistry, East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yi-Lun Ying
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China .,Department of Chemistry, East China University of Science and Technology Shanghai 200237 P. R. China
| | - Wei-Hua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yong-Jing Wan
- School of Information Science and Engineering, East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yi-Tao Long
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China .,Department of Chemistry, East China University of Science and Technology Shanghai 200237 P. R. China
| |
Collapse
|
59
|
Louka A, Zacco E, Temussi PA, Tartaglia GG, Pastore A. RNA as the stone guest of protein aggregation. Nucleic Acids Res 2020; 48:11880-11889. [PMID: 33068411 PMCID: PMC7708036 DOI: 10.1093/nar/gkaa822] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/12/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
The study of prions as infectious aggregates dates several decades. From its original formulation, the definition of a prion has progressively changed to the point that many aggregation-prone proteins are now considered bona fide prions. RNA molecules, not included in the original 'protein-only hypothesis', are also being recognized as important factors contributing to the 'prion behaviour', that implies the transmissibility of an aberrant fold. In particular, an association has recently emerged between aggregation and the assembly of prion-like proteins in RNA-rich complexes, associated with both physiological and pathological events. Here, we discuss the historical rising of the concept of prion-like domains, their relation to RNA and their role in protein aggregation. As a paradigmatic example, we present the case study of TDP-43, an RNA-binding prion-like protein associated with amyotrophic lateral sclerosis. Through this example, we demonstrate how the current definition of prions has incorporated quite different concepts making the meaning of the term richer and more stimulating. An important message that emerges from our analysis is the dual role of RNA in protein aggregation, making RNA, that has been considered for many years a 'silent presence' or the 'stone guest' of protein aggregation, an important component of the process.
Collapse
Affiliation(s)
- Alexandra Louka
- UK Dementia Research Institute at the Maurice Wohl Institute of King's College London, London SE5 9RT, UK
| | - Elsa Zacco
- Center for Human Technologies, Central RNA laboratory, Istituto Italiano di Tecnologia, Genova 16152, Italy
| | - Piero Andrea Temussi
- UK Dementia Research Institute at the Maurice Wohl Institute of King's College London, London SE5 9RT, UK
- University “Federico II’’ Napoli, via Cynthia, Napoli 80100, Italy
| | - Gian Gaetano Tartaglia
- Center for Human Technologies, Central RNA laboratory, Istituto Italiano di Tecnologia, Genova 16152, Italy
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain and ICREA, 23 Passeig Lluıs Companys, Barcelona 08010, Spain
- Charles Darwin department of Biology and Biotechnology, Sapienza University of Rome, Piazzale A. Moro 5, Rome 00185, Italy
| | - Annalisa Pastore
- UK Dementia Research Institute at the Maurice Wohl Institute of King's College London, London SE5 9RT, UK
| |
Collapse
|
60
|
Antibody Fragments as Tools for Elucidating Structure-Toxicity Relationships and for Diagnostic/Therapeutic Targeting of Neurotoxic Amyloid Oligomers. Int J Mol Sci 2020; 21:ijms21238920. [PMID: 33255488 PMCID: PMC7727795 DOI: 10.3390/ijms21238920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
The accumulation of amyloid protein aggregates in tissues is the basis for the onset of diseases known as amyloidoses. Intriguingly, many amyloidoses impact the central nervous system (CNS) and usually are devastating diseases. It is increasingly apparent that neurotoxic soluble oligomers formed by amyloidogenic proteins are the primary molecular drivers of these diseases, making them lucrative diagnostic and therapeutic targets. One promising diagnostic/therapeutic strategy has been the development of antibody fragments against amyloid oligomers. Antibody fragments, such as fragment antigen-binding (Fab), scFv (single chain variable fragments), and VHH (heavy chain variable domain or single-domain antibodies) are an alternative to full-length IgGs as diagnostics and therapeutics for a variety of diseases, mainly because of their increased tissue penetration (lower MW compared to IgG), decreased inflammatory potential (lack of Fc domain), and facile production (low structural complexity). Furthermore, through the use of in vitro-based ligand selection, it has been possible to identify antibody fragments presenting marked conformational selectivity. In this review, we summarize significant reports on antibody fragments selective for oligomers associated with prevalent CNS amyloidoses. We discuss promising results obtained using antibody fragments as both diagnostic and therapeutic agents against these diseases. In addition, the use of antibody fragments, particularly scFv and VHH, in the isolation of unique oligomeric assemblies is discussed as a strategy to unravel conformational moieties responsible for neurotoxicity. We envision that advances in this field may lead to the development of novel oligomer-selective antibody fragments with superior selectivity and, hopefully, good clinical outcomes.
Collapse
|
61
|
The effect of three polyphenols and some other antioxidant substances on amyloid fibril formation by Human cystatin C. Neurochem Int 2020; 140:104806. [DOI: 10.1016/j.neuint.2020.104806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/26/2020] [Accepted: 07/07/2020] [Indexed: 01/24/2023]
|
62
|
Pandey G, Ramakrishnan V. Invasive and non-invasive therapies for Alzheimer's disease and other amyloidosis. Biophys Rev 2020; 12:1175-1186. [PMID: 32930962 PMCID: PMC7575678 DOI: 10.1007/s12551-020-00752-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Advancements in medical science have facilitated in extending human lives. The increased life expectancy, though, has come at a cost. The cases of an aging population suffering from degenerative diseases like Alzheimer's disease (AD) are presently at its all-time high. Amyloidosis disorders such as AD are triggered by an abnormal transition of soluble proteins into their highly ordered aggregated forms. The landscape of amyloidosis treatment remains unchanged, and there is no cure for such disorders. However, an increased understanding of the mechanism of amyloid self-assembly has given hope for a possible therapeutic solution. In this review, we will discuss the current state of molecular and non-molecular options for therapeutic intervention of amyloidosis. We highlight the efficacy of non-invasive physical therapies as possible alternatives to their molecular counterparts. Graphical abstract.
Collapse
Affiliation(s)
- Gaurav Pandey
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, India
| | - Vibin Ramakrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, India.
| |
Collapse
|
63
|
Ghosh P, De P. Modulation of Amyloid Protein Fibrillation by Synthetic Polymers: Recent Advances in the Context of Neurodegenerative Diseases. ACS APPLIED BIO MATERIALS 2020; 3:6598-6625. [DOI: 10.1021/acsabm.0c01021] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Pooja Ghosh
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246 Nadia, West Bengal, India
| | - Priyadarsi De
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246 Nadia, West Bengal, India
| |
Collapse
|
64
|
α-Lactalbumin, Amazing Calcium-Binding Protein. Biomolecules 2020; 10:biom10091210. [PMID: 32825311 PMCID: PMC7565966 DOI: 10.3390/biom10091210] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
α-Lactalbumin (α-LA) is a small (Mr 14,200), acidic (pI 4–5), Ca2+-binding protein. α-LA is a regulatory component of lactose synthase enzyme system functioning in the lactating mammary gland. The protein possesses a single strong Ca2+-binding site, which can also bind Mg2+, Mn2+, Na+, K+, and some other metal cations. It contains several distinct Zn2+-binding sites. Physical properties of α-LA strongly depend on the occupation of its metal binding sites by metal ions. In the absence of bound metal ions, α-LA is in the molten globule-like state. The binding of metal ions, and especially of Ca2+, increases stability of α-LA against the action of heat, various denaturing agents and proteases, while the binding of Zn2+ to the Ca2+-loaded protein decreases its stability and causes its aggregation. At pH 2, the protein is in the classical molten globule state. α-LA can associate with membranes at neutral or slightly acidic pH at physiological temperatures. Depending on external conditions, α-LA can form amyloid fibrils, amorphous aggregates, nanoparticles, and nanotubes. Some of these aggregated states of α-LA can be used in practical applications such as drug delivery to tissues and organs. α-LA and some of its fragments possess bactericidal and antiviral activities. Complexes of partially unfolded α-LA with oleic acid are cytotoxic to various tumor and bacterial cells. α-LA in the cytotoxic complexes plays a role of a delivery carrier of cytotoxic fatty acid molecules into tumor and bacterial cells across the cell membrane. Perhaps in the future the complexes of α-LA with oleic acid will be used for development of new anti-cancer drugs.
Collapse
|
65
|
Caballero AB, Gamez P. Nanochaperone-Based Strategies to Control Protein Aggregation Linked to Conformational Diseases. Angew Chem Int Ed Engl 2020; 60:41-52. [PMID: 32706460 DOI: 10.1002/anie.202007924] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Indexed: 12/14/2022]
Abstract
The generation of highly organized amyloid fibrils is associated with a wide range of conformational pathologies, including primarily neurodegenerative diseases. Such disorders are characterized by misfolded proteins that lose their normal physiological roles and acquire toxicity. Recent findings suggest that proteostasis network impairment may be one of the causes leading to the accumulation and spread of amyloids. These observations are certainly contributing to a new focus in anti-amyloid drug design, whose efforts are so far being centered on single-target approaches aimed at inhibiting amyloid aggregation. Chaperones, known to maintain proteostasis, hence represent interesting targets for the development of novel therapeutics owing to their potential protective role against protein misfolding diseases. In this minireview, research on nanoparticles that can either emulate or help molecular chaperones in recognizing and/or correcting protein misfolding is discussed. The nascent concept of "nanochaperone" may indeed set future directions towards the development of cost-effective, disease-modifying drugs to treat several currently fatal disorders.
Collapse
Affiliation(s)
- Ana B Caballero
- nanoBIC, Departament de Química Inorgànica i Orgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028, Barcelona, Spain
| | - Patrick Gamez
- nanoBIC, Departament de Química Inorgànica i Orgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028, Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, 08010, Barcelona, Spain
| |
Collapse
|
66
|
Caballero AB, Gamez P. Nanochaperone‐Based Strategies to Control Protein Aggregation Linked to Conformational Diseases. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202007924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ana B. Caballero
- nanoBIC Departament de Química Inorgànica i Orgànica Universitat de Barcelona Martí i Franquès, 1–11 08028 Barcelona Spain
- Institute of Nanoscience and Nanotechnology (IN2UB) Universitat de Barcelona 08028 Barcelona Spain
| | - Patrick Gamez
- nanoBIC Departament de Química Inorgànica i Orgànica Universitat de Barcelona Martí i Franquès, 1–11 08028 Barcelona Spain
- Institute of Nanoscience and Nanotechnology (IN2UB) Universitat de Barcelona 08028 Barcelona Spain
- Catalan Institution for Research and Advanced Studies (ICREA) Passeig Lluís Companys 23 08010 Barcelona Spain
| |
Collapse
|
67
|
Monhemi H, Tabaee SS. The effects of mutation and modification on the structure and stability of human lysozyme: A molecular link between carbamylation and atherosclerosis. J Mol Graph Model 2020; 100:107703. [PMID: 32799051 DOI: 10.1016/j.jmgm.2020.107703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/01/2020] [Accepted: 07/18/2020] [Indexed: 01/09/2023]
Abstract
Amino acid mutations in some proteins such as lysozyme lead to genetically disorder variants and adverse pathogenic consequences. Recently, amino acid modifications were known as a risk factor in many related diseases such as uremia and atherosclerosis, showing the importance of these surface-structure changes. Although the structural consequences of the hereditary proteins have been examined extensively, such effects for the protein modifications are known to a lesser extent. One drawback in the examination of protein modifications is hardness in experimental detection of modifications by techniques such as NMR and crystallography. Molecular modeling and simulation can help to understand such phenomena at the molecular levels. It is more rational that the effects of both mutation and modification can be compared in a single protein model. Here, molecular dynamics simulation is used to compare the effects of a disease-related carbamylation modification and an amyloidogenic mutation (D67H) in human lysozyme as a model protein. The results show that the carbamylation adversely effects on the tertiary structure, leading to the similar unfolding pathway to the hereditary amyloidogenic form. The carbamylation leads to the instability of the overall protein conformation, especially on the β-domain, which is a characteristic of hereditary amyloidosis in human lysozymes. The aggregation behaviors of both modified and mutant lysozyme were examined by molecular docking calculations. The results showed that the partially unfolded lysozyme might form tight protein aggregates upon carbamylation similar to the amyloidogenic variant. Both single and all-residues carbamylations impose serious conformational changes to the tertiary structure of lysozyme. It was obtained that carbamylation of lysozyme strongly effects on the stability of N-terminal β-sheet, which can produce a highly unstable conformation. The results of this study not only show the adverse structural consequences of a disease-associated post-translational modification, but it also may be very helpful to understand the molecular basis for many carbamylation-related diseases such as atherosclerosis in ESRD patients. The results show that non-native post-translational modifications may be as structurally important as hereditary mutations.
Collapse
Affiliation(s)
- Hassan Monhemi
- Department of Chemistry, University of Neyshabur, Neyshabur, Iran.
| | - Seyedeh Samaneh Tabaee
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran; Faculty of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
68
|
Söderbom G, Zeng BY. The NLRP3 inflammasome as a bridge between neuro-inflammation in metabolic and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:345-391. [PMID: 32739011 DOI: 10.1016/bs.irn.2020.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Evidence increasingly suggests that type 2 diabetes mellitus (T2DM) is a risk factor for neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases share many pathological processes, including oxidative stress, local inflammation/neuroinflammation and chronic, low-grade (systemic) inflammation, which are exacerbated by aging, a common risk factor for T2DM and NDDs. Here, we focus on the link between chronic inflammation driven by peripheral metabolic disease and how this may impact neurodegeneration in AD and PD. We review the relationship between these common pathological processes in AD and PD from the perspective of the "pro-inflammatory" signaling of the nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeat- (LRR)-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complex. Since the need for effective disease-modifying therapies in T2DM, AD and PD is significant, the relationship between these diseases is important as a positive clinical impact on one may benefit the others. We briefly consider how novel strategies may target neuro-inflammation and provide potential therapies for AD and PD.
Collapse
Affiliation(s)
| | - Bai-Yun Zeng
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
69
|
Broersen K. Milk Processing Affects Structure, Bioavailability and Immunogenicity of β-lactoglobulin. Foods 2020; 9:foods9070874. [PMID: 32635246 PMCID: PMC7404694 DOI: 10.3390/foods9070874] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Bovine milk is subjected to various processing steps to warrant constant quality and consumer safety. One of these steps is pasteurization, which involves the exposure of liquid milk to a high temperature for a limited amount of time. While such heating effectively ameliorates consumer safety concerns mediated by pathogenic bacteria, these conditions also have an impact on one of the main nutritional whey constituents of milk, the protein β-lactoglobulin. As a function of heating, β-lactoglobulin was shown to become increasingly prone to denaturation, aggregation, and lactose conjugation. This review discusses the implications of such heat-induced modifications on digestion and adsorption in the gastro-intestinal tract, and the responses these conformations elicit from the gastro-intestinal immune system.
Collapse
Affiliation(s)
- Kerensa Broersen
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Postbus 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
70
|
Faramarzian M, Bahramikia S. The effects of Quercus brantiiacorn extract on hen egg‐white lysozyme amyloid formation and disassemble amyloid aggregates. J FOOD PROCESS PRES 2020. [DOI: 10.1111/jfpp.14499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
71
|
Simpson LW, Good TA, Leach JB. Protein folding and assembly in confined environments: Implications for protein aggregation in hydrogels and tissues. Biotechnol Adv 2020; 42:107573. [PMID: 32512220 DOI: 10.1016/j.biotechadv.2020.107573] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/03/2020] [Accepted: 05/30/2020] [Indexed: 12/20/2022]
Abstract
In the biological milieu of a cell, soluble crowding molecules and rigid confined environments strongly influence whether the protein is properly folded, intrinsically disordered proteins assemble into distinct phases, or a denatured or aggregated protein species is favored. Such crowding and confinement factors act to exclude solvent volume from the protein molecules, resulting in an increased local protein concentration and decreased protein entropy. A protein's structure is inherently tied to its function. Examples of processes where crowding and confinement may strongly influence protein function include transmembrane protein dimerization, enzymatic activity, assembly of supramolecular structures (e.g., microtubules), nuclear condensates containing transcriptional machinery, protein aggregation in the contexts of disease and protein therapeutics. Historically, most protein structures have been determined from pure, dilute protein solutions or pure crystals. However, these are not the environments in which these proteins function. Thus, there has been an increased emphasis on analyzing protein structure and dynamics in more "in vivo-like" environments. Complex in vitro models using hydrogel scaffolds to study proteins may better mimic features of the in vivo environment. Therefore, analytical techniques need to be optimized for real-time analysis of proteins within hydrogel scaffolds.
Collapse
Affiliation(s)
- Laura W Simpson
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Theresa A Good
- Division of Molecular and Cellular Biosciences, National Science Foundation, 2415 Eisenhower Ave, Alexandria, VA 22314, USA
| | - Jennie B Leach
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
72
|
Meesaragandla B, Karanth S, Janke U, Delcea M. Biopolymer-coated gold nanoparticles inhibit human insulin amyloid fibrillation. Sci Rep 2020; 10:7862. [PMID: 32398693 PMCID: PMC7217893 DOI: 10.1038/s41598-020-64010-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022] Open
Abstract
Deposits of protein misfolding and/or aggregates are a pathological hallmark of amyloid-related diseases. For instance, insulin amyloid fibril deposits have been observed in patients with insulin-dependent diabetes mellitus after insulin administration. Here, we report on the use of AuNPs functionalized with linear- (i.e. dextrin and chitosan) and branched- (i.e. dextran-40 and dextran-10) biopolymers as potential agents to inhibit insulin fibril formation. Our dynamic light scattering analyses showed a size decrease of the amyloid fibrils in the presence of functionalized AuNPs. Circular dichroism spectroscopy as well as enzyme-linked immunosorbent assay data demonstrated that the secondary structural transition from α-helix to β-sheet (which is characteristic for insulin amyloid fibril formation) was significantly suppressed by all biopolymer-coated AuNPs, and in particular, by those functionalized with linear biopolymers. Both transmission electron microscopy and atomic force microscopy analyses showed that the long thick amyloid fibrils formed by insulin alone become shorter, thinner or cluster when incubated with biopolymer-coated AuNPs. Dextrin- and chitosan-coated AuNPs were found to be the best inhibitors of the fibril formation. Based on these results, we propose a mechanism for the inhibition of insulin amyloid fibrils: biopolymer-coated AuNPsstrongly interact with the insulin monomers and inhibit the oligomer formation as well as elongation of the protofibrils.Moreover, cytotoxicity experiments showed that AuNP-insulin amyloid fibrils are less toxic compared to insulin amyloid fibrils alone. Our results suggest that both dextrin- and chitosan-AuNPs could be used as therapeutic agents for the treatment of amyloid-related disorders.
Collapse
Affiliation(s)
- Brahmaiah Meesaragandla
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany.,ZIK HIKE - Zentrum für Innovationskompetenz, Humorale Immunreaktionen bei kardiovaskulären Erkrankungen", Fleischmannstraße 42, 17489, Greifswald, Germany
| | - Sanjai Karanth
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany.,ZIK HIKE - Zentrum für Innovationskompetenz, Humorale Immunreaktionen bei kardiovaskulären Erkrankungen", Fleischmannstraße 42, 17489, Greifswald, Germany
| | - Una Janke
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany.,ZIK HIKE - Zentrum für Innovationskompetenz, Humorale Immunreaktionen bei kardiovaskulären Erkrankungen", Fleischmannstraße 42, 17489, Greifswald, Germany
| | - Mihaela Delcea
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany. .,ZIK HIKE - Zentrum für Innovationskompetenz, Humorale Immunreaktionen bei kardiovaskulären Erkrankungen", Fleischmannstraße 42, 17489, Greifswald, Germany. .,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site, Greifswald, Germany.
| |
Collapse
|
73
|
Dubois V, Serrano D, Zhang X, Seeger S. Structure Analysis of Amyloid Aggregates at Lipid Bilayers by Supercritical Angle Raman Microscopy. Anal Chem 2020; 92:4963-4970. [PMID: 32181651 DOI: 10.1021/acs.analchem.9b05092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The amyloid-β peptide is correlated with Alzheimer's disease and is assumed to cause toxicity by its interaction with the neuron membrane. A custom-made microscope objective based on the supercritical angle technique was developed by our group, which allows investigation of interfacial events by performing surface-sensitive and low-invasive spectroscopy. Applied to Raman spectroscopy, this technique was used to collect information about the structure of polypeptides that interact with a supported lipid bilayer. Notably, the conformation used by amyloid-β(1-40) and amyloid-β(1-42) when interacting directly with or next to the supported lipid bilayer was characterized. We observed two distinct secondary structures, α-helix and β-sheet, which were exhibited by the peptide. These two structures were detected simultaneously. The propensity of the peptide to fold into these structures seemed dependent on both their number of amino acids and their proximity with the supported lipid bilayer. The α-helix structure was observed for amyloid-β(1-42) fragments that were closer to the lipid bilayer. Peptides that were located further away from the bilayer favored the β-sheet structure. Amyloid-β(1-40) was less prone to adopt the α-helix secondary structure.
Collapse
Affiliation(s)
- Valentin Dubois
- Department of Chemistry, University of Zürich, Wintherthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Diana Serrano
- Department of Chemistry, University of Zürich, Wintherthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Xiaotian Zhang
- Department of Chemistry, University of Zürich, Wintherthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Stefan Seeger
- Department of Chemistry, University of Zürich, Wintherthurerstrasse 190, CH-8057 Zürich, Switzerland
| |
Collapse
|
74
|
Islam Z, Ali MH, Popelka A, Mall R, Ullah E, Ponraj J, Kolatkar PR. Probing the fibrillation of lysozyme by nanoscale-infrared spectroscopy. J Biomol Struct Dyn 2020; 39:1481-1490. [PMID: 32131712 DOI: 10.1080/07391102.2020.1734091] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Amyloid fibrillation is the root cause of several neuro as well as non-neurological disorders. Understanding the molecular basis of amyloid aggregate formation is crucial for deciphering various neurodegenerative diseases. In our study, we have examined the lysozyme fibrillation process using nano-infrared spectroscopy (nanoIR). NanoIR enabled us to investigate both structural and chemical characteristics of lysozyme fibrillar species concurrently. The spectroscopic results indicate that lysozyme transformed into a fibrillar structure having mainly parallel β-sheets, with almost no antiparallel β-sheets. Features such as protein stiffness have a good correlation with obtained secondary structural information showing the state of the protein within the fibrillation state. The structural and chemical details were compared with transmission electron microscopy (TEM) and circular dichroism (CD). We have utilized nanoIR and measured infrared spectra to characterize lysozyme amyloid fibril structures in terms of morphology, molecular structure, secondary structure content, stability, and size of the cross-β core. We have shown that the use of nanoIR can complement other biophysical studies to analyze the aggregation process and is particularly useful for studying proteins involved in aggregation to help in designing molecules against amyloid aggregation. Specifically, the nanoIR spectra afford higher resolution information and a characteristic fingerprint for determining states of aggregation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zeyaul Islam
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Mohamed H Ali
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Anton Popelka
- Center for Advanced Materials (CAM), Qatar University, Doha, Qatar
| | - Raghvendra Mall
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
| | - Ehsan Ullah
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University, Doha, Qatar
| | - Janarthanan Ponraj
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University, Doha, Qatar
| | - Prasanna R Kolatkar
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
75
|
Ratha BN, Kar RK, Bednarikova Z, Gazova Z, Kotler SA, Raha S, De S, Maiti NC, Bhunia A. Molecular Details of a Salt Bridge and Its Role in Insulin Fibrillation by NMR and Raman Spectroscopic Analysis. J Phys Chem B 2020; 124:1125-1136. [PMID: 31958230 DOI: 10.1021/acs.jpcb.9b10349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Insulin, a simple polypeptide hormone with huge biological importance, has long been known to self-assemble in vitro and form amyloid-like fibrillar aggregates. Utilizing high-resolution NMR, Raman spectroscopy, and computational analysis, we demonstrate that the fluctuation of the carboxyl terminal (C-ter) residues of the insulin B-chain plays a key role in the growth phase of insulin aggregation. By comparing the insulin sourced from bovine, human, and the modified glargine (GI), we observed reduced aggregation propensity in the GI variant, resulting from two additional Arg residues at its C-ter. NMR analysis showed atomic contacts and residue-specific interactions, particularly the salt bridge and H-bond formed among the C-ter residues Arg31B, Lys29B, and Glu4A. These inter-residue interactions were reflected in strong nuclear Overhauser effects among Arg31BδH-Glu4AδH and Lys29BδHs-Glu4AδH in GI, as well as the associated downfield chemical shift of several A-chain amino terminal (N-ter) residues. The two additional Arg residues of GI, Arg31B and Arg32B, enhanced the stability of the GI native structure by strengthening the Arg31B, Lys29B, and Glu4A salt bridge, thus reducing extensive thermal distortion and fluctuation of the terminal residues. The high stability of the salt bridge retards tertiary collapse, a crucial biochemical event for oligomerization and subsequent fibril formation. Circular dichroism and Raman spectroscopic measurement also suggest slow structural distortion in the early phase of the aggregation of GI because of the restricted mobility of the C-ter residues as explained by NMR. In addition, the structural and dynamic parameters derived from molecular dynamics simulations of insulin variants highlight the role of residue-specific contacts in aggregation and amyloid-like fibril formation.
Collapse
Affiliation(s)
- Bhisma N Ratha
- Department of Biophysics , Bose Institute , P-1/12 CIT Scheme VII (M) , Kolkata 700054 , India
| | - Rajiv K Kar
- Department of Biophysics , Bose Institute , P-1/12 CIT Scheme VII (M) , Kolkata 700054 , India
| | - Zuzana Bednarikova
- Department of Biophysics , Institute of Experimental Physics Slovak Academy of Sciences , Kosice 040 01 , Slovakia
| | - Zuzana Gazova
- Department of Biophysics , Institute of Experimental Physics Slovak Academy of Sciences , Kosice 040 01 , Slovakia
| | - Samuel A Kotler
- National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland 20850 , United States
| | - Sreyan Raha
- Department of Physics , Bose Institute , 93/1 APC Road , Kolkata 700009 , India
| | - Soumya De
- School of Bioscience , IIT Kharagpur , Kharagpur 721302 , India
| | - Nakul C Maiti
- Division Structural Biology and Bioinformatics , CSIR-Indian Institute of Chemical Biology , Kolkata 700032 , India
| | - Anirban Bhunia
- Department of Biophysics , Bose Institute , P-1/12 CIT Scheme VII (M) , Kolkata 700054 , India
| |
Collapse
|
76
|
Pariary R, Ghosh B, Bednarikova Z, Varnava KG, Ratha BN, Raha S, Bhattacharyya D, Gazova Z, Sarojini V, Mandal AK, Bhunia A. Targeted inhibition of amyloidogenesis using a non-toxic, serum stable strategically designed cyclic peptide with therapeutic implications. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140378. [PMID: 32032759 DOI: 10.1016/j.bbapap.2020.140378] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/24/2020] [Accepted: 01/31/2020] [Indexed: 10/25/2022]
Abstract
Amyloidogenic disorders are currently rising as a global health issue, prompting more and more studies dedicated to the development of effective targeted therapeutics. The innate affinity of these amyloidogenic proteins towards the biomembranes adds further complexities to the systems. Our previous studies have shown that biologically active peptides can effectively target amyloidogenesis serving as an efficient therapeutic alternative in several amyloidogenic disorders. The structural uniqueness of the PWWP motif in the de novo designed heptapeptide, KR7 (KPWWPRR-NH2) was demonstrated to target insulin fiber elongation specifically. By working on insulin, an important model system in amyloidogenic studies, we gained several mechanistic insights into the inhibitory actions at the protein-peptide interface. Here, we report a second-generation non-toxic and serum stable cyclic peptide, based primarily on the PWWP motif that resulted in complete inhibition of insulin fibrillation both in the presence and absence of the model membranes. Using both low- and high-resolution spectroscopic techniques, we could delineate the specific mechanism of inhibition, at atomistic resolution. Our studies put forward an effective therapeutic intervention that redirects the default aggregation kinetics towards off-pathway fibrillation. Based on the promising results, this novel cyclic peptide can be considered an excellent lead to design pharmaceutical molecules against amyloidogenesis.
Collapse
Affiliation(s)
- Ranit Pariary
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata, 700054, India
| | - Baijayanti Ghosh
- Department of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata, 700054, India
| | - Zuzana Bednarikova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Kosice, Slovakia
| | - Kyriakos Gabriel Varnava
- School of Chemical Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Bhisma N Ratha
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata, 700054, India
| | - Sreyan Raha
- Department of Physics, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Dipita Bhattacharyya
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata, 700054, India
| | - Zuzana Gazova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Kosice, Slovakia
| | - Vijayalekshmi Sarojini
- School of Chemical Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Atin K Mandal
- Department of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata, 700054, India
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata, 700054, India.
| |
Collapse
|
77
|
Alshamleh I, Krause N, Richter C, Kurrle N, Serve H, Günther UL, Schwalbe H. Real-Time NMR Spectroscopy for Studying Metabolism. Angew Chem Int Ed Engl 2020; 59:2304-2308. [PMID: 31730253 PMCID: PMC7004128 DOI: 10.1002/anie.201912919] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Current metabolomics approaches utilize cellular metabolite extracts, are destructive, and require high cell numbers. We introduce here an approach that enables the monitoring of cellular metabolism at lower cell numbers by observing the consumption/production of different metabolites over several kinetic data points of up to 48 hours. Our approach does not influence cellular viability, as we optimized the cellular matrix in comparison to other materials used in a variety of in-cell NMR spectroscopy experiments. We are able to monitor real-time metabolism of primary patient cells, which are extremely sensitive to external stress. Measurements are set up in an interleaved manner with short acquisition times (approximately 7 minutes per sample), which allows the monitoring of up to 15 patient samples simultaneously. Further, we implemented our approach for performing tracer-based assays. Our approach will be important not only in the metabolomics fields, but also in individualized diagnostics.
Collapse
Affiliation(s)
- Islam Alshamleh
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Str. 760438FrankfurtGermany
| | - Nina Krause
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Str. 760438FrankfurtGermany
| | - Christian Richter
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Str. 760438FrankfurtGermany
| | - Nina Kurrle
- German Cancer Consortium (DKTK) and DKFZ69120HeidelbergGermany
- Department of Medicine 2, Hematology/OncologyGoethe University60590Frankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)60590Frankfurt am MainGermany
| | - Hubert Serve
- German Cancer Consortium (DKTK) and DKFZ69120HeidelbergGermany
- Department of Medicine 2, Hematology/OncologyGoethe University60590Frankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)60590Frankfurt am MainGermany
| | - Ulrich L. Günther
- Institute of Chemistry and MetabolomicsUniversity of LuebeckRatzeburger Allee 16023562LuebeckGermany
| | - Harald Schwalbe
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University FrankfurtMax-von-Laue Str. 760438FrankfurtGermany
| |
Collapse
|
78
|
Bernson D, Mecinovic A, Abed MT, Limé F, Jageland P, Palmlöf M, Esbjörner EK. Amyloid formation of bovine insulin is retarded in moderately acidic pH and by addition of short-chain alcohols. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2020; 49:145-153. [PMID: 31901953 PMCID: PMC7069927 DOI: 10.1007/s00249-019-01420-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 11/24/2022]
Abstract
Protein aggregation and amyloid formation are associated with multiple human diseases, but are also a problem in protein production. Understanding how aggregation can be modulated is therefore of importance in both medical and industrial contexts. We have used bovine insulin as a model protein to explore how amyloid formation is affected by buffer pH and by the addition of short-chain alcohols. We find that bovine insulin forms amyloid fibrils, albeit with different rates and resulting fibril morphologies, across a wide pH range (2–7). At pH 4.0, bovine insulin displayed relatively low aggregation propensity in combination with high solubility; this condition was therefore chosen as basis for further exploration of how bovine insulin’s native state can be stabilized in the presence of short-chain alcohols that are relevant because of their common use as eluents in industrial-scale chromatography purification. We found that ethanol and isopropanol are efficient modulators of bovine insulin aggregation, providing a three to four times retardation of the aggregation kinetics at 30–35% (vol/vol) concentration; we attribute this to the formation of oligomers, which we detected by AFM. We discuss this effect in terms of reduced solvent polarity and show, by circular dichroism recordings, that a concomitant change in α-helical packing of the insulin monomer occurs in ethanol. Our results extend current knowledge of how insulin aggregates, and may, although bovine insulin serves as a simplistic model, provide insights into how buffers and additives can be fine-tuned in industrial production of proteins in general and pharmaceutical insulin in particular.
Collapse
Affiliation(s)
- David Bernson
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 412 96, Gothenburg, Sweden
| | - Almedina Mecinovic
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 412 96, Gothenburg, Sweden
| | - Md Tuhin Abed
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 412 96, Gothenburg, Sweden
| | - Fredrik Limé
- Nouyron Pulp and Performance Chemicals AB, Separation Products, 445 80, Bohus, Sweden
| | - Per Jageland
- Nouyron Pulp and Performance Chemicals AB, Separation Products, 445 80, Bohus, Sweden
| | - Magnus Palmlöf
- Nouyron Pulp and Performance Chemicals AB, Separation Products, 445 80, Bohus, Sweden
| | - Elin K Esbjörner
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 412 96, Gothenburg, Sweden.
| |
Collapse
|
79
|
Alshamleh I, Krause N, Richter C, Kurrle N, Serve H, Günther UL, Schwalbe H. Real‐Time NMR Spectroscopy for Studying Metabolism. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201912919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Islam Alshamleh
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University Frankfurt Max-von-Laue Str. 7 60438 Frankfurt Germany
| | - Nina Krause
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University Frankfurt Max-von-Laue Str. 7 60438 Frankfurt Germany
| | - Christian Richter
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University Frankfurt Max-von-Laue Str. 7 60438 Frankfurt Germany
| | - Nina Kurrle
- German Cancer Consortium (DKTK) and DKFZ 69120 Heidelberg Germany
- Department of Medicine 2, Hematology/OncologyGoethe University 60590 Frankfurt am Main Germany
- Frankfurt Cancer Institute (FCI) 60590 Frankfurt am Main Germany
| | - Hubert Serve
- German Cancer Consortium (DKTK) and DKFZ 69120 Heidelberg Germany
- Department of Medicine 2, Hematology/OncologyGoethe University 60590 Frankfurt am Main Germany
- Frankfurt Cancer Institute (FCI) 60590 Frankfurt am Main Germany
| | - Ulrich L. Günther
- Institute of Chemistry and MetabolomicsUniversity of Luebeck Ratzeburger Allee 160 23562 Luebeck Germany
| | - Harald Schwalbe
- Institute of Organic Chemistry and Chemical BiologyCenter for Biomolecular Magnetic Resonance (BMRZ)Johann Wolfgang Goethe-University Frankfurt Max-von-Laue Str. 7 60438 Frankfurt Germany
| |
Collapse
|
80
|
Poulson BG, Szczepski K, Lachowicz JI, Jaremko L, Emwas AH, Jaremko M. Aggregation of biologically important peptides and proteins: inhibition or acceleration depending on protein and metal ion concentrations. RSC Adv 2019; 10:215-227. [PMID: 35492549 PMCID: PMC9047971 DOI: 10.1039/c9ra09350h] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 12/14/2019] [Indexed: 01/03/2023] Open
Abstract
The process of aggregation of proteins and peptides is dependent on the concentration of proteins, and the rate of aggregation can be altered by the presence of metal ions, but this dependence is not always a straightforward relationship. In general, aggregation does not occur under normal physiological conditions, yet it can be induced in the presence of certain metal ions. However, the extent of the influence of metal ion interactions on protein aggregation has not yet been fully comprehended. A consensus has thus been difficult to reach because the acceleration/inhibition of the aggregation of proteins in the presence of metal ions depends on several factors such as pH and the concentration of the aggregated proteins involved as well as metal concentration level of metal ions. Metal ions, like Cu2+, Zn2+, Pb2+ etc. may either accelerate or inhibit aggregation simply because the experimental conditions affect the behavior of biomolecules. It is clear that understanding the relationship between metal ion concentration and protein aggregation will prove useful for future scientific applications. This review focuses on the dependence of the aggregation of selected important biomolecules (peptides and proteins) on metal ion concentrations. We review proteins that are prone to aggregation, the result of which can cause serious neurodegenerative disorders. Furthering our understanding of the relationship between metal ion concentration and protein aggregation will prove useful for future scientific applications, such as finding therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Benjamin Gabriel Poulson
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Kacper Szczepski
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Joanna Izabela Lachowicz
- Department of Medical Sciences and Public Health, University of Cagliari, Cittadella Universitaria 09042 Monserrato Italy
| | - Lukasz Jaremko
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| | - Mariusz Jaremko
- Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900 Saudi Arabia
| |
Collapse
|
81
|
Protein Microgels from Amyloid Fibril Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1174:223-263. [PMID: 31713201 DOI: 10.1007/978-981-13-9791-2_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Nanofibrillar forms of amyloidogenic proteins were initially discovered in the context of protein misfolding and disease but have more recently been found at the origin of key biological functionality in many naturally occurring functional materials, such as adhesives and biofilm coatings. Their physiological roles in nature reflect their great strength and stability, which has led to the exploration of their use as the basis of artificial protein-based functional materials. Particularly for biomedical applications, they represent attractive building blocks for the development of, for instance, drug carrier agents due to their inherent biocompatibility and biodegradability. Furthermore, the propensity of proteins to self-assemble into amyloid fibrils can be exploited under microconfinement, afforded by droplet microfluidic techniques. This approach allows the generation of multi-scale functional microgels that can host biological additives and can be designed to incorporate additional functionality, such as to aid targeted drug delivery.
Collapse
|
82
|
Bocedi A, Cattani G, Gambardella G, Ticconi S, Cozzolino F, Di Fusco O, Pucci P, Ricci G. Ultra-Rapid Glutathionylation of Ribonuclease: Is this the Real Incipit of its Oxidative Folding? Int J Mol Sci 2019; 20:ijms20215440. [PMID: 31683668 PMCID: PMC6862303 DOI: 10.3390/ijms20215440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 11/16/2022] Open
Abstract
Many details of oxidative folding of proteins remain obscure, in particular, the role of oxidized glutathione (GSSG). This study reveals some unknown aspects. When a reduced ribonuclease A refolds in the presence of GSSG, most of its eight cysteines accomplish a very fast glutathionylation. In particular, one single cysteine, identified as Cys95 by mass spectrometry, displays 3600 times higher reactivity when compared with an unperturbed protein cysteine. Furthermore, the other five cysteines show 40-50 times higher reactivity toward GSSG. This phenomenon is partially due to a low pKa value of most of these cysteines (average pKa = 7.9), but the occurrence of a reversible GSSG-ribonuclease complex (KD = 0.12 mM) is reasonably responsible for the extraordinary hyper-reactivity of Cys95. Neither hyper-reactivity nor some protein-disulfide complexes have been found by reacting a reduced ribonuclease with other natural disulfides i.e., cystine, cystamine, and homocystine. Hyper-reactivity of all cysteines was observed toward 5,5'-dithiobis-(2-nitrobenzoic acid). Given that GSSG is present in high concentrations in the endoplasmic reticulum, this property may shed light on the early step of its oxidative folding. The ultra-rapid glutathionylation of cysteines, only devoted to form disulfides, is a novel property of the molten globule status of the ribonuclease.
Collapse
Affiliation(s)
- Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Giada Cattani
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Giorgia Gambardella
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Silvia Ticconi
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Flora Cozzolino
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II", 80126 Naples, Italy.
| | - Ornella Di Fusco
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II", 80126 Naples, Italy.
| | - Piero Pucci
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II", 80126 Naples, Italy.
| | - Giorgio Ricci
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
83
|
Navalkar A, Ghosh S, Pandey S, Paul A, Datta D, Maji SK. Prion-like p53 Amyloids in Cancer. Biochemistry 2019; 59:146-155. [DOI: 10.1021/acs.biochem.9b00796] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Ambuja Navalkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Saikat Ghosh
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Satyaprakash Pandey
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Ajoy Paul
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Debalina Datta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, India 400076
| |
Collapse
|
84
|
Mroczko B, Groblewska M, Litman-Zawadzka A. The Role of Protein Misfolding and Tau Oligomers (TauOs) in Alzheimer's Disease (AD). Int J Mol Sci 2019; 20:E4661. [PMID: 31547024 PMCID: PMC6802364 DOI: 10.3390/ijms20194661] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 11/25/2022] Open
Abstract
Although the causative role of the accumulation of amyloid β 1-42 (Aβ42) deposits in the pathogenesis of Alzheimer's disease (AD) has been under debate for many years, it is supposed that the toxicity soluble oligomers of Tau protein (TauOs) might be also the pathogenic factor acting on the initial stages of this disease. Therefore, we performed a thorough search for literature pertaining to our investigation via the MEDLINE/PubMed database. It was shown that soluble TauOs, especially granular forms, may be the most toxic form of this protein. Hyperphosphorylated TauOs can reduce the number of synapses by missorting into axonal compartments of neurons other than axon. Furthermore, soluble TauOs may be also responsible for seeding Tau pathology within AD brains, with probable link to AβOs toxicity. Additionally, the concentrations of TauOs in the cerebrospinal fluid (CSF) and plasma of AD patients were higher than in non-demented controls, and revealed a negative correlation with mini-mental state examination (MMSE) scores. It was postulated that adding the measurements of TauOs to the panel of CSF biomarkers could improve the diagnosis of AD.
Collapse
Affiliation(s)
- Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland.
- Department of Biochemical Diagnostics, University Hospital of Białystok, 15-269 Białystok, Poland.
| | - Magdalena Groblewska
- Department of Biochemical Diagnostics, University Hospital of Białystok, 15-269 Białystok, Poland.
| | - Ala Litman-Zawadzka
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland.
| |
Collapse
|
85
|
Siddiqi MK, Malik S, Majid N, Alam P, Khan RH. Cytotoxic species in amyloid-associated diseases: Oligomers or mature fibrils. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 118:333-369. [PMID: 31928731 DOI: 10.1016/bs.apcsb.2019.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyloid diseases especially, Alzheimer's disease (AD), is characterized by an imbalance between the production and clearance of amyloid-β (Aβ) species. Amyloidogenic proteins or peptides can transform structurally from monomers into β-stranded fibrils via multiple oligomeric states. Among various amyloid species, structured oligomers are proposed to be more toxic than fibrils; however, the identification of amyloid oligomers has been challenging due to their heterogeneous and metastable nature. Multiple techniques have recently helped in better understanding of oligomer's assembly details and structural properties. Moreover, some progress on elucidating the mechanisms of oligomer-triggered toxicity has been made. Based on the collection of current findings, there is growing consensus that control of toxic amyloid oligomers could be a valid approach to regulate amyloid-associated toxicity, which could advance development of new diagnostics and therapeutics for amyloid-related diseases. In this review, we have described the recent scenario of amyloid diseases with a great deal of information about the recent understanding of oligomers' assembly, structural properties, and toxicity. Also comprehensive details have been provided to differentiate the degree of toxicity associated with prefibrillar aggregates.
Collapse
Affiliation(s)
| | - Sadia Malik
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Nabeela Majid
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Parvez Alam
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
86
|
Lysozyme encapsulated gold nanoclusters for probing the early stage of lysozyme aggregation under acidic conditions. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 197:111540. [DOI: 10.1016/j.jphotobiol.2019.111540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/02/2019] [Accepted: 06/21/2019] [Indexed: 12/22/2022]
|
87
|
Es-haghi A, Jahedi Moghaddam M, Shahpasand K. Role of Pre-molten Globule Structure in Protein Amyloid Fibril Formation. AVICENNA JOURNAL OF MEDICAL BIOCHEMISTRY 2019. [DOI: 10.34172/ajmb.2019.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The conversion of a protein from its native conformation to the pathogenic form is a critical event in the pathogenesis of several neurodegenerative disorders such as Alzheimer’s (AD), Parkinson’s, and Huntington’s diseases, along with type II diabetic mellitus. Although there are several reports on the mechanism of protein aggregation, the actual conformation playing a part in the pathogenicity is yet unclear. Accordingly, the present study summarizes the early pathogenic conformation resulting in several protein aggregations. It is well-documented that a pre-molten globule (MG) structure appears at the early stages of some proteins. Pre-MG is one of the intermediate structures, which is formed during some protein unfolding processes. In addition, it is shown that the pre-molten structure is more flexible than the mature MG one and thus, protein easily rearranges to form amyloid fibrils in this conformation. Therefore, protein aggregation is halted by preventing the pre-MG structure. The strategy of protein aggregation prevention has profound implications in fighting the devastating disorder.
Collapse
Affiliation(s)
- Ali Es-haghi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Centre of Education, Culture and Research, Tehran, Iran
| |
Collapse
|
88
|
|
89
|
Samui S, Biswas S, Roy K, Deb I, Naskar J. Engineering of Supramolecular β-Sheet and Nontoxic Amyloid Fibrils from Synthetic Oligopeptides Containing γ-Aminobutyric Acid as the N-Terminal Residue. ACS Chem Neurosci 2019; 10:2915-2918. [PMID: 31082196 DOI: 10.1021/acschemneuro.9b00119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Here we demonstrate that three synthetic tripeptides containing conformationally flexible γ-aminobutyric acid (γ-Abu) as the N-terminal residue form supramolecular β-sheet and nanofibrillar aggregates upon self-association in aqueous medium. Congo red and thioflavin T binding study establish that these nanofibrillar aggregates are amyloidogenic in nature. The MTT cell survival assay suggests that these amyloid-like nanofibrillar aggregates are nontoxic toward cultured Neuro 2A cells. Interestingly, none of these tripeptides bear sequence identity with any amyloid forming proteins or peptides; however upon self-association, they form supramolecular β-sheet and amyloid-like nanofibrils those are nontoxic in nature. The results highlight the self-assembling nature of the conformationally flexible peptides in aqueous environment and support the hypothesis that amyloid formation is the intrinsic property of the polypeptide chain. Also the cytotoxicity is not predictive from amyloid fibril formation alone. Such nontoxic amyloid fibrils can be exploited in future to design functional biomaterials for various biomedical applications.
Collapse
Affiliation(s)
- Satyabrata Samui
- Department of Biochemistry & Biophysics, University of Kalyani, Nadia, WB 741235, India
| | - Soumi Biswas
- Department of Biochemistry & Biophysics, University of Kalyani, Nadia, WB 741235, India
| | - Kaninika Roy
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Rd., Kolkata, 700019, India
| | - Ishani Deb
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Rd., Kolkata, 700019, India
| | - Jishu Naskar
- Department of Biochemistry & Biophysics, University of Kalyani, Nadia, WB 741235, India
| |
Collapse
|
90
|
Chaari A. Molecular chaperones biochemistry and role in neurodegenerative diseases. Int J Biol Macromol 2019; 131:396-411. [DOI: 10.1016/j.ijbiomac.2019.02.148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023]
|
91
|
Trigo D, Nadais A, da Cruz e Silva OA. Unravelling protein aggregation as an ageing related process or a neuropathological response. Ageing Res Rev 2019; 51:67-77. [PMID: 30763619 DOI: 10.1016/j.arr.2019.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/07/2019] [Accepted: 02/07/2019] [Indexed: 12/18/2022]
Abstract
Protein aggregation is normally associated with amyloidosis, namely motor neurone, Alzheimer's, Parkinson's or prion diseases. However, recent results have unveiled a concept of gradual increase of protein aggregation associated with the ageing process, apparently not necessarily associated with pathological conditions. Given that protein aggregation is sufficient to activate stress-response and inflammation, impairing protein synthesis and quality control mechanisms, the former is assumed to negatively affect cellular metabolism and behaviour. In this review the state of the art in protein aggregation research is discussed, namely the relationship between pathology and proteostasis. The role of pathology and ageing in overriding protein quality-control mechanisms, and consequently, the effect of these faulty cellular processes on pathological and healthy ageing, are also addressed.
Collapse
|
92
|
Perween S, Abidi M, Faizy AF, Moinuddin. Post-translational modifications on glycated plasma fibrinogen: A physicochemical insight. Int J Biol Macromol 2019; 126:1201-1212. [DOI: 10.1016/j.ijbiomac.2019.01.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/11/2018] [Accepted: 01/04/2019] [Indexed: 12/29/2022]
|
93
|
Crowder induced structural modulation of a multi-domain protein during its early stages of aggregation: A FRET-based and protein solvation study. Int J Biol Macromol 2019; 127:563-574. [DOI: 10.1016/j.ijbiomac.2019.01.054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 01/02/2019] [Accepted: 01/11/2019] [Indexed: 12/31/2022]
|
94
|
Release of Pharmaceutical Peptides in an Aggregated State: Using Fibrillar Polymorphism to Modulate Release Levels. COLLOIDS AND INTERFACES 2019. [DOI: 10.3390/colloids3010042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Traditional approaches to achieve sustained delivery of pharmaceutical peptides traditionally use co-excipients (e.g., microspheres and hydrogels). Here, we investigate the release of an amyloidogenic glucagon analogue (3474) from an aggregated state and the influence of surfactants on this process. The formulation of peptide 3474 in dodecyl maltoside (DDM), rhamnolipid (RL), and sophorolipid (SL) led to faster fibrillation. When the aggregates were subjected to multiple cycles of release by repeated resuspension in fresh buffer, the kinetics of the release of soluble peptide 3474 from different surfactant aggregates all followed a simple exponential decay fit, with half-lives of 5–18 min and relatively constant levels of release in each cycle. However, different amounts of peptide are released from different aggregates, ranging from 0.015 mg/mL (3475-buffer) up to 0.03 mg/mL (3474-DDM), with 3474-buffer and 3474-RL in between. In addition to higher release levels, 3474-DDM aggregates showed a different amyloid FTIR structure, compared to 3474-RL and 3474-SL aggregates and a faster rate of degradation by proteinase K. This demonstrates that the stability of organized peptide aggregates can be modulated to achieve differences in release of soluble peptides, thus coupling aggregate polymorphism to differential release profiles. We achieved aggregate polymorphism by the addition of different surfactants, but polymorphism may also be reached through other approaches, including different excipients as well as changes in pH and salinity, providing a versatile handle to control release profiles.
Collapse
|
95
|
Chandel TI, Masroor A, Siddiqi MK, Siddique IA, Jahan I, Ali M, Nayeem SM, Uversky VN, Khan RH. Molecular basis of the inhibition and disaggregation of thermally-induced amyloid fibrils of human serum albumin by an anti-Parkinson's drug, benserazide hydrochloride. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2018.12.127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
96
|
Joseph SK, Kuritz N, Yahel E, Lapshina N, Rosenman G, Natan A. Proton-Transfer-Induced Fluorescence in Self-Assembled Short Peptides. J Phys Chem A 2019; 123:1758-1765. [DOI: 10.1021/acs.jpca.8b09183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sijo K. Joseph
- Department of Physical Electronics, Tel-Aviv University, Tel-Aviv 69978, Israel
- The Sackler Center for Computational Molecular and Materials Science, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Natalia Kuritz
- Department of Physical Electronics, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Eldad Yahel
- Department of Physical Electronics, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Nadezda Lapshina
- Department of Physical Electronics, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Gil Rosenman
- Department of Physical Electronics, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Amir Natan
- Department of Physical Electronics, Tel-Aviv University, Tel-Aviv 69978, Israel
- The Sackler Center for Computational Molecular and Materials Science, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
97
|
Alkudaisi N, Russell BA, Jachimska B, Birch DJS, Chen Y. Detecting lysozyme unfolding via the fluorescence of lysozyme encapsulated gold nanoclusters. J Mater Chem B 2019; 7:1167-1175. [PMID: 32254785 DOI: 10.1039/c9tb00009g] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Protein misfolding plays a critical role in the manifestation of amyloidosis type diseases. Therefore, understanding protein unfolding and the ability to track protein unfolding in a dynamic manner are of considerable interest. Fluorescence-based techniques are powerful tools for gaining real-time information about the local environmental conditions of a probe on the nanoscale. Fluorescent gold nanoclusters (AuNCs) are a new type of fluorescent probes which are <2 nm in diameter, incredibly robust and offer highly sensitive, wavelength tuneable emission. Their small size minimises intrusion and makes AuNCs ideal for studying protein dynamics. Lysozyme has previously been used to encapsulate AuNCs. The unfolding dynamics of lysozyme under different environmental conditions have been well-studied and being an amyloid type protein makes lysozyme an ideal candidate for encapsulating AuNCs in order to test their sensitivity to protein unfolding. In this study, we tracked the fluorescence characteristics of AuNCs encapsulated in lysozyme while inducing protein unfolding using urea, sodium dodecyl sulphate (SDS) and elevated temperature and compared them to complimentary circular dichroism spectra. It is found that AuNC fluorescence emission is quenched upon induced protein unfolding either due to a decrease in Forster Resonance Energy Transfer (FRET) efficiency between tryptophan and AuNCs or solvent exposure of the AuNC. Fluorescence lifetime measurements confirmed quenching to be collisional via oxygen dissolved in a solution which increases as the AuNC was exposed to the solvent during unfolding. Moreover, the longer decay component τ1 was observed to decrease as the protein unfolded, due to the increased collisional quenching. It is suggested that AuNC sensitivity to solvent exposure might be utilised in the future as a new approach to studying and possibly even detecting amyloidosis type diseases.
Collapse
Affiliation(s)
- Nora Alkudaisi
- Department of Physics, SUPA, University of Strathclyde, John Anderson Building, 107 Rottenrow, Glasgow G4 0NG, UK.
| | | | | | | | | |
Collapse
|
98
|
Bonito-Oliva A, Schedin-Weiss S, Younesi SS, Tiiman A, Adura C, Paknejad N, Brendel M, Romin Y, Parchem RJ, Graff C, Vukojević V, Tjernberg LO, Terenius L, Winblad B, Sakmar TP, Graham WV. Conformation-specific antibodies against multiple amyloid protofibril species from a single amyloid immunogen. J Cell Mol Med 2019; 23:2103-2114. [PMID: 30663210 PMCID: PMC6378190 DOI: 10.1111/jcmm.14119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/28/2018] [Accepted: 12/07/2018] [Indexed: 12/01/2022] Open
Abstract
We engineered and employed a chaperone‐like amyloid‐binding protein Nucleobindin 1 (NUCB1) to stabilize human islet amyloid polypeptide (hIAPP) protofibrils for use as immunogen in mice. We obtained multiple monoclonal antibody (mAb) clones that were reactive against hIAPP protofibrils. A secondary screen was carried out to identify clones that cross‐reacted with amyloid beta‐peptide (Aβ42) protofibrils, but not with Aβ40 monomers. These mAbs were further characterized in several in vitro assays, in immunohistological studies of a mouse model of Alzheimer's disease (AD) and in AD patient brain tissue. We show that mAbs obtained by immunizing mice with the NUCB1‐hIAPP complex cross‐react with Aβ42, specifically targeting protofibrils and inhibiting their further aggregation. In line with conformation‐specific binding, the mAbs appear to react with an intracellular antigen in diseased tissue, but not with amyloid plaques. We hypothesize that the mAbs we describe here recognize a secondary or quaternary structural epitope that is common to multiple amyloid protofibrils. In summary, we report a method to create mAbs that are conformation‐sensitive and sequence‐independent and can target more than one type of protofibril species.
Collapse
Affiliation(s)
- Alessandra Bonito-Oliva
- Laboratory of Chemical Biology & Signal Transduction, The Rockefeller University, New York City, New York
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - Shahab S Younesi
- Department of Neuroscience, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas
| | - Ann Tiiman
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carolina Adura
- High Throughput and Spectroscopy Resource Center, The Rockefeller University, New York City, New York
| | - Navid Paknejad
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York City, New York
| | - Matt Brendel
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York City, New York
| | - Yevgeniy Romin
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York City, New York
| | - Ronald J Parchem
- Department of Neuroscience, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas
| | - Caroline Graff
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - Thomas P Sakmar
- Laboratory of Chemical Biology & Signal Transduction, The Rockefeller University, New York City, New York.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - W Vallen Graham
- Laboratory of Chemical Biology & Signal Transduction, The Rockefeller University, New York City, New York
| |
Collapse
|
99
|
Röder K, Joseph JA, Husic BE, Wales DJ. Energy Landscapes for Proteins: From Single Funnels to Multifunctional Systems. ADVANCED THEORY AND SIMULATIONS 2019. [DOI: 10.1002/adts.201800175] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Konstantin Röder
- Department of ChemistryUniversity of CambridgeLensfield Road CB2 1EW Cambridge UK
| | - Jerelle A. Joseph
- Department of ChemistryUniversity of CambridgeLensfield Road CB2 1EW Cambridge UK
| | - Brooke E. Husic
- Department of ChemistryUniversity of CambridgeLensfield Road CB2 1EW Cambridge UK
| | - David J. Wales
- Department of ChemistryUniversity of CambridgeLensfield Road CB2 1EW Cambridge UK
| |
Collapse
|
100
|
Fitzgerald BW. The physiology of impenetrable skin: Colossus of the X-Men. ADVANCES IN PHYSIOLOGY EDUCATION 2018; 42:529-540. [PMID: 30192188 DOI: 10.1152/advan.00107.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The X-Men are an ensemble of superheroes whose powers are associated with the X-Gene, a mutant genetic factor. The powers exhibited by each character differ and are dependent on how the X-Gene has modified their individual genomes. For instance, Wolverine possesses regenerative healing, Storm can control local weather systems, and Colossus can create an impenetrable "organic steel" layer around his body. Thanks to the establishment of the superhero genre in modern cinema, audiences are familiar with Colossus from films such as X-Men: Days of Future Past and Deadpool. While attaining this power might be attractive to many people, there are innumerate scientific obstacles to be overcome to replicate this "organic steel" layer. Due to its unique combination of high strength and flexibility, a graphene-based layer might be a more realistic material for Colossus' impenetrable skin and would also address a number of physiological issues associated with an "organic steel" layer. The actualization of this layer would depend on complex processes associated with protein folding, protein self-assembly, and changing the structure of his skin. In the classroom, Colossus can foster a multidisciplinary learning environment where concepts in physiology can overlap with topics in physics, engineering, and materials science. Just like other superheroes, Colossus can also be used to promote scientific content in outreach for the general public.
Collapse
Affiliation(s)
- Barry W Fitzgerald
- Intensified Reaction and Separation Systems, Department of Process and Energy, Delft University of Technology , Delft , The Netherlands
| |
Collapse
|