1
|
González Díaz A, Cataldi R, Mannini B, Vendruscolo M. Preparation and Characterization of Zn(II)-Stabilized Aβ 42 Oligomers. ACS Chem Neurosci 2024; 15:2586-2599. [PMID: 38979921 PMCID: PMC11258685 DOI: 10.1021/acschemneuro.4c00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
Aβ oligomers are being investigated as cytotoxic agents in Alzheimer's disease (AD). Because of their transient nature and conformational heterogeneity, the relationship between the structure and activity of these oligomers is still poorly understood. Hence, methods for stabilizing Aβ oligomeric species relevant to AD are needed to uncover the structural determinants of their cytotoxicity. Here, we build on the observation that metal ions and metabolites have been shown to interact with Aβ, influencing its aggregation and stabilizing its oligomeric species. We thus developed a method that uses zinc ions, Zn(II), to stabilize oligomers produced by the 42-residue form of Aβ (Aβ42), which is dysregulated in AD. These Aβ42-Zn(II) oligomers are small in size, spanning the 10-30 nm range, stable at physiological temperature, and with a broad toxic profile in human neuroblastoma cells. These oligomers offer a tool to study the mechanisms of toxicity of Aβ oligomers in cellular and animal AD models.
Collapse
Affiliation(s)
- Alicia González Díaz
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Rodrigo Cataldi
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Benedetta Mannini
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
- Department
of Experimental and Clinical Biomedical Sciences Mario Serio, University
of Florence, 50134 Florence, Italy
| | - Michele Vendruscolo
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
2
|
McCalpin SD, Khemtemourian L, Suladze S, Ivanova MI, Reif B, Ramamoorthy A. Zinc and pH modulate the ability of insulin to inhibit aggregation of islet amyloid polypeptide. Commun Biol 2024; 7:776. [PMID: 38937578 PMCID: PMC11211420 DOI: 10.1038/s42003-024-06388-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/27/2024] [Indexed: 06/29/2024] Open
Abstract
Aggregation of the human islet amyloid polypeptide (hIAPP) contributes to the development and progression of Type 2 Diabetes (T2D). hIAPP aggregates within a few hours at few micromolar concentration in vitro but exists at millimolar concentrations in vivo. Natively occurring inhibitors of hIAPP aggregation might therefore provide a model for drug design against amyloid formation associated with T2D. Here, we describe the combined ability of low pH, zinc, and insulin to inhibit hIAPP fibrillation. Insulin dose-dependently slows hIAPP aggregation near neutral pH but had less effect on the aggregation kinetics at acidic pH. We determine that insulin alters hIAPP aggregation in two manners. First, insulin diverts the aggregation pathway to large nonfibrillar aggregates with ThT-positive molecular structure, rather than to amyloid fibrils. Second, soluble insulin suppresses hIAPP dimer formation, which is an important early aggregation event. Further, we observe that zinc significantly modulates the inhibition of hIAPP aggregation by insulin. We hypothesize that this effect arose from controlling the oligomeric state of insulin and show that hIAPP interacts more strongly with monomeric than oligomeric insulin.
Collapse
Affiliation(s)
- Samuel D McCalpin
- Biophysics Program, University of Michigan, Arbor, MI, 48109, USA
- Department of Chemistry, University of Michigan, Arbor, MI, 48109, USA
| | - Lucie Khemtemourian
- Institute of Chemistry and Biology of Membranes and Nanoobjects (CBMN), CNRS - UMR 5248, Institut Polytechnique Bordeaux, University of Bordeaux, 33600, Pessac, France
| | - Saba Suladze
- Bayerisches NMR Zentrum (BNMRZ) at the Department of Biosciences, School of Natural Sciences, Technische Universität München, Munich, Germany
- Helmholtz-Zentrum München (HMGU), Deutsches Forschungszentrum für Gesundheit und Umwelt, Institute of Structural Biology (STB), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Magdalena I Ivanova
- Biophysics Program, University of Michigan, Arbor, MI, 48109, USA
- Department of Neurology, University of Michigan, Arbor, MI, 48109, USA
- Michigan Neuroscience Institute, University of Michigan, Arbor, MI, 48109, USA
| | - Bernd Reif
- Bayerisches NMR Zentrum (BNMRZ) at the Department of Biosciences, School of Natural Sciences, Technische Universität München, Munich, Germany
- Helmholtz-Zentrum München (HMGU), Deutsches Forschungszentrum für Gesundheit und Umwelt, Institute of Structural Biology (STB), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Ayyalusamy Ramamoorthy
- Biophysics Program, University of Michigan, Arbor, MI, 48109, USA.
- Department of Chemistry, University of Michigan, Arbor, MI, 48109, USA.
- Department of Neurology, University of Michigan, Arbor, MI, 48109, USA.
- Michigan Neuroscience Institute, University of Michigan, Arbor, MI, 48109, USA.
- Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- National High Magnetic Field Laboratory, Department of Chemical and Biomedical Engineering, Institute of Molecular Biophysics, Neuroscience, Florida State University, Tallahassee, FL, 32310, USA.
| |
Collapse
|
3
|
Meleleo D, Cibelli G, Valenzano A, Mastrodonato M, Mallamaci R. The Effect of Calcium Ions on hIAPP Channel Activity: Possible Implications in T2DM. MEMBRANES 2023; 13:878. [PMID: 37999364 PMCID: PMC10673357 DOI: 10.3390/membranes13110878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023]
Abstract
The calcium ion (Ca2+) has been linked to type 2 diabetes mellitus (T2DM), although the role of Ca2+ in this disorder is the subject of intense investigation. Serum Ca2+ dyshomeostasis is associated with the development of insulin resistance, reduced insulin sensitivity, and impaired glucose tolerance. However, the molecular mechanisms involving Ca2+ ions in pancreatic β-cell loss and subsequently in T2DM remain poorly understood. Implicated in the decline in β-cell functions are aggregates of human islet amyloid polypeptide (hIAPP), a small peptide secreted by β-cells that shows a strong tendency to self-aggregate into β-sheet-rich aggregates that evolve toward the formation of amyloid deposits and mature fibrils. The soluble oligomers of hIAPP can permeabilize the cell membrane by interacting with bilayer lipids. Our study aimed to evaluate the effect of Ca2+ on the ability of the peptide to incorporate and form ion channels in zwitterionic planar lipid membranes (PLMs) composed of palmitoyl-oleoyl-phosphatidylcholine (POPC) and on the aggregation process of hIAPP molecules in solution. Our results may help to clarify the link between Ca2+ ions, hIAPP peptide, and consequently the pathophysiology of T2DM.
Collapse
Affiliation(s)
- Daniela Meleleo
- Department of Science of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.C.); (A.V.)
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.C.); (A.V.)
| | - Maria Mastrodonato
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.M.); (R.M.)
| | - Rosanna Mallamaci
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.M.); (R.M.)
| |
Collapse
|
4
|
Li MH, Zhang X, London E, Raleigh DP. Impact of Ca 2+ on membrane catalyzed IAPP amyloid formation and IAPP induced vesicle leakage. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184161. [PMID: 37121365 PMCID: PMC10735052 DOI: 10.1016/j.bbamem.2023.184161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/10/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023]
Abstract
Human islet amyloid polypeptide (hIAPP, also known as amylin) is a 37 amino acid pancreatic polypeptide hormone that plays a role in regulating glucose levels, but forms pancreatic amyloid in type-2 diabetes. The process of amyloid formation by hIAPP contributes to β-cell death in the disease. Multiple mechanisms of hIAPP induced toxicity of β-cells have been proposed including disruption of cellular membranes. However, the nature of hIAPP membrane interactions and the effect of ions and other molecules on hIAPP membrane interactions are not fully understood. Many studies have used model membranes with a high content of anionic lipids, often POPS, however the concentration of anionic lipids in the β-cell plasma membrane is low. Here we study the concentration dependent effect of Ca2+ (0 to 50 mM) on hIAPP membrane interactions using large unilamellar vesicles (LUVs) with anionic lipid content ranging from 0 to 50 mol%. We find that Ca2+ does not effectively inhibit hIAPP amyloid formation and hIAPP induced membrane leakage from binary LUVs with a low percentage of POPS, but has a greater effect on LUVs with a high percentage of POPS. Mg2+ had very similar effects, and the effects of Ca2+ and Mg2+ can be largely rationalized by the neutralization of POPS charge. The implications for hIAPP-membrane interactions are discussed.
Collapse
Affiliation(s)
- Ming-Hao Li
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Xiaoxue Zhang
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, United States
| | - Erwin London
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY 11794, United States; Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, United States; Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, United States.
| | - Daniel P Raleigh
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY 11794, United States; Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, United States; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States.
| |
Collapse
|
5
|
Zinc and iron dynamics in human islet amyloid polypeptide-induced diabetes mouse model. Sci Rep 2023; 13:3484. [PMID: 36922503 PMCID: PMC10017767 DOI: 10.1038/s41598-023-30498-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Metal homeostasis is tightly regulated in cells and organisms, and its disturbance is frequently observed in some diseases such as neurodegenerative diseases and metabolic disorders. Previous studies suggest that zinc and iron are necessary for the normal functions of pancreatic β cells. However, the distribution of elements in normal conditions and the pathophysiological significance of dysregulated elements in the islet in diabetic conditions have remained unclear. In this study, to investigate the dynamics of elements in the pancreatic islets of a diabetic mouse model expressing human islet amyloid polypeptide (hIAPP): hIAPP transgenic (hIAPP-Tg) mice, we performed imaging analysis of elements using synchrotron scanning X-ray fluorescence microscopy and quantitative analysis of elements using inductively coupled plasma mass spectrometry. We found that in the islets, zinc significantly decreased in the early stage of diabetes, while iron gradually decreased concurrently with the increase in blood glucose levels of hIAPP-Tg mice. Notably, when zinc and/or iron were decreased in the islets of hIAPP-Tg mice, dysregulation of glucose-stimulated mitochondrial respiration was observed. Our findings may contribute to clarifying the roles of zinc and iron in islet functions under pathophysiological diabetic conditions.
Collapse
|
6
|
Suh JM, Kim M, Yoo J, Han J, Paulina C, Lim MH. Intercommunication between metal ions and amyloidogenic peptides or proteins in protein misfolding disorders. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
7
|
Evidence of the different effect of mercury and cadmium on the hIAPP aggregation process. Biophys Chem 2022; 290:106880. [DOI: 10.1016/j.bpc.2022.106880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/22/2022]
|
8
|
Abstract
Amyloids are organized suprastructural polypeptide arrangements. The prevalence of amyloid-related processes of pathophysiological relevance has been linked to aging-related degenerative diseases. Besides the role of genetic polymorphisms on the relative risk of amyloid diseases, the contributions of nongenetic ontogenic cluster of factors remain elusive. In recent decades, mounting evidences have been suggesting the role of essential micronutrients, in particular transition metals, in the regulation of amyloidogenic processes, both directly (such as binding to amyloid proteins) or indirectly (such as regulating regulatory partners, processing enzymes, and membrane transporters). The features of transition metals as regulatory cofactors of amyloid proteins and the consequences of metal dyshomeostasis in triggering amyloidogenic processes, as well as the evidences showing amelioration of symptoms by dietary supplementation, suggest an exaptative role of metals in regulating amyloid pathways. The self- and cross-talk replicative nature of these amyloid processes along with their systemic distribution support the concept of their metastatic nature. The role of amyloidosis as nutrient sensors would act as intra- and transgenerational epigenetic metabolic programming factors determining health span and life span, viability, which could participate as an evolutive selective pressure.
Collapse
Affiliation(s)
- Luís Maurício T R Lima
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory for Macromolecules (LAMAC-DIMAV), National Institute of Metrology, Quality and Technology - INMETRO, Duque de Caxias, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tháyna Sisnande
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
10
|
Abstract
Amyloid diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and type 2 diabetes (T2D) are characterized by accumulation of misfolded proteins' species, e.g., oligomers and fibrils. The formation of these species occurs via self-assemble of the misfolded proteins in a process which is named "aggregation." It is known that essential divalent metal ions initiate the aggregation of these misfolded proteins, and that specific concentrations of these metal ions may be implicated in the pathology of amyloid diseases. This chapter focuses on the effects of two of the most common divalent metal ions in the brain-Zn2+ and Cu2+, and while Zn2+ ion is known as a metal that is release from the pancreas. Specifically, the spotlight of this chapter illustrates recent computational molecular modelling studies that investigate the effect of the concentrations of metal ions on aggregation of the misfolded proteins amylin, amyloid β, and α-synuclein. The challenges for computational molecular modeling and future perspectives are discussed.
Collapse
Affiliation(s)
- Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
| |
Collapse
|
11
|
Khemtemourian L, Antoniciello F, Sahoo BR, Decossas M, Lecomte S, Ramamoorthy A. Investigation of the effects of two major secretory granules components, insulin and zinc, on human-IAPP amyloid aggregation and membrane damage. Chem Phys Lipids 2021; 237:105083. [PMID: 33887213 DOI: 10.1016/j.chemphyslip.2021.105083] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/11/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
Human islet amyloid polypeptide (hIAPP) is a highly amyloidogenic peptide found in pancreatic islets of type-2 diabetes (T2D) patients. Under certain conditions, hIAPP is able to form amyloid fibrils that play a role in the progression of T2D. hIAPP is synthesized in the β-cell of the pancreas and stored in the secretory granules before being released into the extracellular compartment. It has been suggested that natural stabilizing agents, such as insulin or zinc present in the secretory granules with hIAPP could prevent hIAPP fibril formation. The difference in the amino acid sequences of IAPP among species strongly correlates with amyloidogenicity and toxicity. The residue histidine at position 18 is known to be important in modulating the fibril formation, membrane leakage and toxicity. In this study, we have synthesized four analogues of hIAPP (H18R-IAPP, H18K-IAPP, H18A-IAPP and H18E-IAPP) and characterized their aggregation with either insulin or zinc in order to determine the effect of the residue-18 on the insulin-IAPP and zinc-IAPP interactions using a variety of biophysical experiments including thioflavin-T fluorescence, transmission electron microscopy imaging, circular dichroism, and NMR spectroscopy. We show that insulin reduced hIAPP fibril formation both in solution and in the presence of membrane and hIAPP-membrane damage and that the interactions are somewhat mediated by the residue-18. In addition, our results reveal that zinc affects the process of hIAPP fibril formation in solution but not in the presence of membrane. Our results indicate that the nature of the residue-18 is important for zinc binding. Based on this observation, we hypothesize that zinc binds to the residues in the N-terminal region of hIAPP, which is not accessible in the presence of membrane due to its strong interaction with lipids.
Collapse
Affiliation(s)
| | | | - Bikash R Sahoo
- Biophysics Program, Department of Chemistry, Biomedical Engineering, and Macromolecular Science and Engineering, The University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Marion Decossas
- CBMN, CNRS UMR 5248, IPB, Univ. Bordeaux, F-33600 Pessac, France
| | - Sophie Lecomte
- CBMN, CNRS UMR 5248, IPB, Univ. Bordeaux, F-33600 Pessac, France
| | - Ayyalusamy Ramamoorthy
- Biophysics Program, Department of Chemistry, Biomedical Engineering, and Macromolecular Science and Engineering, The University of Michigan, Ann Arbor, MI 48109-1055, USA.
| |
Collapse
|
12
|
Ferguson MQ, DeRosa MC. Optimized experimental pre-treatment strategy for temporary inhibition of islet amyloid polypeptide aggregation. Biochem Biophys Rep 2021; 26:100964. [PMID: 33912690 PMCID: PMC8063701 DOI: 10.1016/j.bbrep.2021.100964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 11/24/2022] Open
Abstract
Islet amyloid polypeptide (IAPP) is a neuroendocrine hormone from pancreatic β-cells. Misfolded, aggregated IAPP is believed to be toxic to islet cells and amyloid deposits in the pancreas are pathological hallmarks of type 2 diabetes. Rapid fibrillization of this peptide makes it difficult to study in its soluble form, impeding a better understanding of its role. In this study, a variety of popular pretreatment methods were tested for their ability to delay aggregation of IAPP, including solutions of hexafluoroisopropanol, sodium hydroxide, hydrochloric acid, phosphate buffered saline, ammonium hydroxide, as well as tris buffer at different pH and containing either calcium (II), zinc (II), or iron (II). Aggregation was assessed using the thioflavin T fluorescence assay as well as by transmission electron microscopy. Tris buffer at pH 8.1 containing Zn(II) was found to have the best balance of temporary inhibition of aggregation and biological relevance.
Collapse
Affiliation(s)
- Madison Q Ferguson
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Maria C DeRosa
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| |
Collapse
|
13
|
Nguyen PH, Ramamoorthy A, Sahoo BR, Zheng J, Faller P, Straub JE, Dominguez L, Shea JE, Dokholyan NV, De Simone A, Ma B, Nussinov R, Najafi S, Ngo ST, Loquet A, Chiricotto M, Ganguly P, McCarty J, Li MS, Hall C, Wang Y, Miller Y, Melchionna S, Habenstein B, Timr S, Chen J, Hnath B, Strodel B, Kayed R, Lesné S, Wei G, Sterpone F, Doig AJ, Derreumaux P. Amyloid Oligomers: A Joint Experimental/Computational Perspective on Alzheimer's Disease, Parkinson's Disease, Type II Diabetes, and Amyotrophic Lateral Sclerosis. Chem Rev 2021; 121:2545-2647. [PMID: 33543942 PMCID: PMC8836097 DOI: 10.1021/acs.chemrev.0c01122] [Citation(s) in RCA: 455] [Impact Index Per Article: 113.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein misfolding and aggregation is observed in many amyloidogenic diseases affecting either the central nervous system or a variety of peripheral tissues. Structural and dynamic characterization of all species along the pathways from monomers to fibrils is challenging by experimental and computational means because they involve intrinsically disordered proteins in most diseases. Yet understanding how amyloid species become toxic is the challenge in developing a treatment for these diseases. Here we review what computer, in vitro, in vivo, and pharmacological experiments tell us about the accumulation and deposition of the oligomers of the (Aβ, tau), α-synuclein, IAPP, and superoxide dismutase 1 proteins, which have been the mainstream concept underlying Alzheimer's disease (AD), Parkinson's disease (PD), type II diabetes (T2D), and amyotrophic lateral sclerosis (ALS) research, respectively, for many years.
Collapse
Affiliation(s)
- Phuong H Nguyen
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Bikash R Sahoo
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Jie Zheng
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Peter Faller
- Institut de Chimie, UMR 7177, CNRS-Université de Strasbourg, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - John E Straub
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Laura Dominguez
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Joan-Emma Shea
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - Nikolay V Dokholyan
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
- Department of Chemistry, and Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Alfonso De Simone
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
- Molecular Biology, University of Naples Federico II, Naples 80138, Italy
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Saeed Najafi
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - Son Tung Ngo
- Laboratory of Theoretical and Computational Biophysics & Faculty of Applied Sciences, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
| | - Antoine Loquet
- Institute of Chemistry & Biology of Membranes & Nanoobjects, (UMR5248 CBMN), CNRS, Université Bordeaux, Institut Européen de Chimie et Biologie, 33600 Pessac, France
| | - Mara Chiricotto
- Department of Chemical Engineering and Analytical Science, University of Manchester, Manchester M13 9PL, U.K
| | - Pritam Ganguly
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - James McCarty
- Chemistry Department, Western Washington University, Bellingham, Washington 98225, United States
| | - Mai Suan Li
- Institute for Computational Science and Technology, SBI Building, Quang Trung Software City, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City 700000, Vietnam
- Institute of Physics, Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Carol Hall
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695-7905, United States
| | - Yiming Wang
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695-7905, United States
| | - Yifat Miller
- Department of Chemistry and The Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | | | - Birgit Habenstein
- Institute of Chemistry & Biology of Membranes & Nanoobjects, (UMR5248 CBMN), CNRS, Université Bordeaux, Institut Européen de Chimie et Biologie, 33600 Pessac, France
| | - Stepan Timr
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Jiaxing Chen
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Brianna Hnath
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, and Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Sylvain Lesné
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Science, Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200438, China
| | - Fabio Sterpone
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Andrew J Doig
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Philippe Derreumaux
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
- Laboratory of Theoretical Chemistry, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
| |
Collapse
|
14
|
Milardi D, Gazit E, Radford SE, Xu Y, Gallardo RU, Caflisch A, Westermark GT, Westermark P, Rosa CL, Ramamoorthy A. Proteostasis of Islet Amyloid Polypeptide: A Molecular Perspective of Risk Factors and Protective Strategies for Type II Diabetes. Chem Rev 2021; 121:1845-1893. [PMID: 33427465 PMCID: PMC10317076 DOI: 10.1021/acs.chemrev.0c00981] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The possible link between hIAPP accumulation and β-cell death in diabetic patients has inspired numerous studies focusing on amyloid structures and aggregation pathways of this hormone. Recent studies have reported on the importance of early oligomeric intermediates, the many roles of their interactions with lipid membrane, pH, insulin, and zinc on the mechanism of aggregation of hIAPP. The challenges posed by the transient nature of amyloid oligomers, their structural heterogeneity, and the complex nature of their interaction with lipid membranes have resulted in the development of a wide range of biophysical and chemical approaches to characterize the aggregation process. While the cellular processes and factors activating hIAPP-mediated cytotoxicity are still not clear, it has recently been suggested that its impaired turnover and cellular processing by proteasome and autophagy may contribute significantly toward toxic hIAPP accumulation and, eventually, β-cell death. Therefore, studies focusing on the restoration of hIAPP proteostasis may represent a promising arena for the design of effective therapies. In this review we discuss the current knowledge of the structures and pathology associated with hIAPP self-assembly and point out the opportunities for therapy that a detailed biochemical, biophysical, and cellular understanding of its aggregation may unveil.
Collapse
Affiliation(s)
- Danilo Milardi
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Via P. Gaifami 18, 95126 Catania, Italy
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Yong Xu
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Rodrigo U Gallardo
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zürich, Zürich CH-8057, Switzerland
| | - Gunilla T Westermark
- Department of Medical Cell Biology, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Per Westermark
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Carmelo La Rosa
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Ayyalusamy Ramamoorthy
- Biophysics, Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan 41809-1055, United States
| |
Collapse
|
15
|
Ben-Shushan S, Miller Y. Insulin fibrillation control by specific zinc binding sites. Inorg Chem Front 2021. [DOI: 10.1039/d1qi01054a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein, we present for the first time a study that identifies the morphology of full-length insulin fibrils in the absence and in the presence of Zn2+ ions.
Collapse
Affiliation(s)
- Shira Ben-Shushan
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beér Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beér-Sheva 84105, Israel
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beér Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beér-Sheva 84105, Israel
| |
Collapse
|
16
|
Wang Y, Meng F, Lu T, Wang C, Li F. Regulation of divalent metal ions to the aggregation and membrane damage of human islet amyloid polypeptide oligomers. RSC Adv 2021; 11:12815-12825. [PMID: 35423832 PMCID: PMC8697352 DOI: 10.1039/d1ra00354b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/21/2021] [Indexed: 11/21/2022] Open
Abstract
The accumulation of human islet amyloid polypeptide (hIAPP) on the surface of pancreatic β cells is closely related to the death of the cells. Divalent metal ions play a significant role in the cytotoxicity of hIAPP. In this study, we examined the roles played by the divalent metal ions of zinc, copper and calcium in the aggregation of both hIAPP18-27 fragment and full-length hIAPP and the ability of their oligomers to damage the membrane of POPC/POPG 4 : 1 LUVs using the ThT fluorescence, TEM, AFM, CD, ANS binding fluorescence and dye leakage experiments. We prepared metal-free and metal-associated oligomers that are similar in size and aggregate slowly using the short peptide and confirmed that the ability of the peptide oligomers to damage the lipid membrane is reduced by the binding to the metal ions, which is closely linked to the reducing hydrophobic exposure of the metal-associated oligomers. The study on the full-length hIAPP showed that the observed membrane damage induced by hIAPP oligomers is either mitigated at a peptide-to-metal ratio of 1 : 0.33 or aggravated at a peptide-to-metal ratio of 1 : 1 in the presence of Zn(ii) and Cu(ii), while the surface hydrophobicity of hIAPP oligomers was reduced at both peptide-to-metal ratios. The observed results of the membrane damage were attributed to the counteraction between a decrease in the disruptive ability of metal-associated oligomer species and an increase in the quantity of oligomers promoted by the binding of the metal ions to hIAPP oligomers. The former could play a predominant role in reducing the membrane damage at a peptide-to-metal ratio of 1 : 0.33, while the latter could play a predominant role in enhancing the membrane damage at a peptide-to-metal ratio of 1 : 1. This study shows that an enhanced membrane damage could be caused by the oligomer species with a decreased instead of an increased disruptive ability, given that the abundance of the oligomer species is high enough. Their is a counteraction between a decrease in the disruptive ability of metal-associated oligomer species and an increase in the quantity of oligomers promoted by the metal binding in the activity of hIAPP induced membrane damage.![]()
Collapse
Affiliation(s)
- Yajie Wang
- State Key Laboratory of Supramolecular Structure and Materials
- Jilin University
- Changchun 130012
- P. R. China
| | - Feihong Meng
- State Key Laboratory of Supramolecular Structure and Materials
- Jilin University
- Changchun 130012
- P. R. China
| | - Tong Lu
- State Key Laboratory of Supramolecular Structure and Materials
- Jilin University
- Changchun 130012
- P. R. China
| | - Chunyun Wang
- State Key Laboratory of Supramolecular Structure and Materials
- Jilin University
- Changchun 130012
- P. R. China
| | - Fei Li
- State Key Laboratory of Supramolecular Structure and Materials
- Jilin University
- Changchun 130012
- P. R. China
| |
Collapse
|
17
|
Saghir AE, Farrugia G, Vassallo N. The human islet amyloid polypeptide in protein misfolding disorders: Mechanisms of aggregation and interaction with biomembranes. Chem Phys Lipids 2020; 234:105010. [PMID: 33227292 DOI: 10.1016/j.chemphyslip.2020.105010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/06/2020] [Accepted: 11/09/2020] [Indexed: 02/09/2023]
Abstract
Human islet amyloid polypeptide (hIAPP), otherwise known as amylin, is a 37-residue peptide hormone which is reported to be a common factor in protein misfolding disorders such as type-2 diabetes mellitus, Alzheimer's disease and Parkinson's disease, due to deposition of insoluble hIAPP amyloid in the pancreas and brain. Multiple studies point to the importance of the peptide's interaction with biological membranes and the cytotoxicity of hIAPP species. Here, we discuss the aggregation pathways of hIAPP amyloid fibril formation and focus on the complex interplay between membrane-mediated assembly of hIAPP and the associated mechanisms of membrane damage caused by the peptide species. Mitochondrial membranes, which are unique in their lipid composition, are proposed as prime targets for the early intracellular formation of hIAPP toxic entities. We suggest that future studies should include more physiologically-relevant and in-cell studies to allow a more accurate model of in vivo interactions. Finally, we underscore an urgent need for developing effective therapeutic strategies aimed at hindering hIAPP-phospholipid interactions.
Collapse
Affiliation(s)
- Adam El Saghir
- Dept. of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Gianluca Farrugia
- Dept. of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Neville Vassallo
- Dept. of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta.
| |
Collapse
|
18
|
Magrì A, Tabbì G, Di Natale G, La Mendola D, Pietropaolo A, Zoroddu MA, Peana M, Rizzarelli E. Zinc Interactions with a Soluble Mutated Rat Amylin to Mimic Whole Human Amylin: An Experimental and Simulation Approach to Understand Stoichiometry, Speciation and Coordination of the Metal Complexes. Chemistry 2020; 26:13072-13084. [DOI: 10.1002/chem.202002114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Indexed: 01/27/2023]
Affiliation(s)
- Antonio Magrì
- Consiglio Nazionale delle Ricerche Istituto di Cristallografia Via P. Gaifami 18 95126 Catania Italy
| | - Giovanni Tabbì
- Consiglio Nazionale delle Ricerche Istituto di Cristallografia Via P. Gaifami 18 95126 Catania Italy
| | - Giuseppe Di Natale
- Consiglio Nazionale delle Ricerche Istituto di Cristallografia Via P. Gaifami 18 95126 Catania Italy
| | - Diego La Mendola
- Dipartimento di Farmacia Università di Pisa Via Bonanno Pisano, 6 56126 Pisa Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei, Metalli nei Sistemi Biologici (CIRCMSB) Via Celso Ulpiani 27 70126 Bari Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute Università “Magna Graecia” di Catanzaro Campus Universitario, Viale Europa 88100 Catanzaro Italy
| | | | - Massimiliano Peana
- Dipartimento di Chimica e Farmacia University of Sassari Via Vienna 2 07100 Sassari Italy
| | - Enrico Rizzarelli
- Consiglio Nazionale delle Ricerche Istituto di Cristallografia Via P. Gaifami 18 95126 Catania Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei, Metalli nei Sistemi Biologici (CIRCMSB) Via Celso Ulpiani 27 70126 Bari Italy
- Dipartimento di Scienze Chimiche Università degli Studi di Catania Viale A. Doria 6 95125 Catania Italy
| |
Collapse
|
19
|
Vane EW, He S, Maibaum L, Nath A. Rapid Formation of Peptide/Lipid Coaggregates by the Amyloidogenic Seminal Peptide PAP 248-286. Biophys J 2020; 119:924-938. [PMID: 32814060 PMCID: PMC7474197 DOI: 10.1016/j.bpj.2020.07.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/14/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022] Open
Abstract
Protein/lipid coassembly is an understudied phenomenon that is important to the function of antimicrobial peptides as well as the pathological effects of amyloid. Here, we study the coassembly process of PAP248-286, a seminal peptide that displays both amyloid-forming and antimicrobial activity. PAP248-286 is a peptide fragment of prostatic acid phosphatase and has been reported to form amyloid fibrils, known as semen-derived enhancer of viral infection (SEVI), that enhance the viral infectivity of human immunodeficiency virus. We find that in addition to forming amyloid, PAP248-286 much more readily assembles with lipid vesicles into peptide/lipid coaggregates that resemble amyloid fibrils in some important ways but are a distinct species. The formation of these PAP248-286/lipid coaggregates, which we term "messicles," is controlled by the peptide:lipid (P:L) ratio and by the lipid composition. The optimal P:L ratio is around 1:10, and at least 70% anionic lipid is required for coaggregate formation. Once formed, messicles are not disrupted by subsequent changes in P:L ratio. We propose that messicles form through a polyvalent assembly mechanism, in which a critical surface density of PAP248-286 on liposomes enables peptide-mediated particle bridging into larger species. Even at ∼50-fold lower PAP248-286 concentrations, messicles form at least 10-fold faster than amyloid fibrils. It is therefore possible that some or all of the biological activities assigned to SEVI, the amyloid form of PAP248-286, could instead be attributed to a PAP248-286/lipid coaggregate. More broadly speaking, this work could provide a potential framework for the discovery and characterization of nonamyloid peptide/lipid coaggregates by other amyloid-forming proteins and antimicrobial peptides.
Collapse
Affiliation(s)
- Eleanor W Vane
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington; Biological Physics, Structure and Design Program, University of Washington, Seattle, Washington
| | - Shushan He
- Department of Chemistry, University of Washington, Seattle, Washington
| | - Lutz Maibaum
- Department of Chemistry, University of Washington, Seattle, Washington
| | - Abhinav Nath
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington; Biological Physics, Structure and Design Program, University of Washington, Seattle, Washington.
| |
Collapse
|
20
|
Amyloidogenic Intrinsically Disordered Proteins: New Insights into Their Self-Assembly and Their Interaction with Membranes. Life (Basel) 2020; 10:life10080144. [PMID: 32784399 PMCID: PMC7459996 DOI: 10.3390/life10080144] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/02/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022] Open
Abstract
Aβ, IAPP, α-synuclein, and prion proteins belong to the amyloidogenic intrinsically disordered proteins’ family; indeed, they lack well defined secondary and tertiary structures. It is generally acknowledged that they are involved, respectively, in Alzheimer’s, Type II Diabetes Mellitus, Parkinson’s, and Creutzfeldt–Jakob’s diseases. The molecular mechanism of toxicity is under intense debate, as many hypotheses concerning the involvement of the amyloid and the toxic oligomers have been proposed. However, the main role is represented by the interplay of protein and the cell membrane. Thus, the understanding of the interaction mechanism at the molecular level is crucial to shed light on the dynamics driving this phenomenon. There are plenty of factors influencing the interaction as mentioned above, however, the overall view is made trickier by the apparent irreproducibility and inconsistency of the data reported in the literature. Here, we contextualized this topic in a historical, and even more importantly, in a future perspective. We introduce two novel insights: the chemical equilibrium, always established in the aqueous phase between the free and the membrane phospholipids, as mediators of protein-transport into the core of the bilayer, and the symmetry-breaking of oligomeric aggregates forming an alternating array of partially ordered and disordered monomers.
Collapse
|
21
|
Sisnande T, Lima CK, da Silva DC, Beninatto TM, Alves NL, Amaral MJ, Miranda-Alves L, Lima LMTR. Dietary zinc restriction promotes degeneration of the endocrine pancreas in mice. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165675. [PMID: 31927001 DOI: 10.1016/j.bbadis.2020.165675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/25/2022]
Abstract
Zinc is a key component of several proteins, interacting with the pancreatic hormones insulin and amylin. The role of zinc in insulin oligomerization and crystallinity is well established, although the effects of dietary zinc restriction on both energetic metabolism and β-pancreatic hormonemia and morphology remain unexplored. Here we report the effects of dietary zinc restriction on the endocrine pancreas and metabolic phenotype of mice. Nontransgenic male Swiss mice were fed a low-zinc or control diet for 4 weeks after weanling. Growth, glycemia, insulinemia and amylinemia were lower and pancreatic islets were smaller in the intervention group despite the preserved insulin crystallinity in secretory granules. We found strong immunostaining for insulin, amylin and oligomers in apoptotic pancreatic islet. High production of β-pancreatic hormones in zinc-restricted animals counteracted the reduced islet size caused by apoptosis. These data suggest that zinc deficiency is sufficient to promote islet β-cell hormonal disruption and degeneration.
Collapse
Affiliation(s)
- Tháyna Sisnande
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Cleverton K Lima
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Dayana Cabral da Silva
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Thayana Moulin Beninatto
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Natália Leão Alves
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Mariana J Amaral
- Hospital Universitário Clementino Fraga Filho, Federal University of Rio de Janeiro - UFRJ, CCS, Ilha do Fundão, 21941-902 Rio de Janeiro, RJ, Brazil.
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Luís Maurício T R Lima
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Laboratory for Macromolecules (LAMAC-DIMAV), National Institute of Metrology, Quality and Technology - INMETRO, Avenida Nossa Senhora das Graças, 50 - Xerém, Duque de Caxias, Rio de Janeiro 25250-020, RJ, Brazil.
| |
Collapse
|
22
|
Altamirano-Bustamante MM, Altamirano-Bustamante NF, Larralde-Laborde M, Lara-Martínez R, Leyva-García E, Garrido-Magaña E, Rojas G, Jiménez-García LF, Revilla-Monsalve C, Altamirano P, Calzada-León R. Unpacking the aggregation-oligomerization-fibrillization process of naturally-occurring hIAPP amyloid oligomers isolated directly from sera of children with obesity or diabetes mellitus. Sci Rep 2019; 9:18465. [PMID: 31804529 PMCID: PMC6895187 DOI: 10.1038/s41598-019-54570-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
The formation of amyloid oligomers and fibrils of the human islet amyloid polypeptide (hIAPP) has been linked with β- cell failure and death which causes the onset, progression, and comorbidities of diabetes. We begin to unpack the aggregation-oligomerization-fibrillization process of these oligomers taken from sera of pediatric patients. The naturally occurring or real hIAPP (not synthetic) amyloid oligomers (RIAO) were successfully isolated, we demonstrated the presence of homo (dodecamers, hexamers, and trimers) and hetero-RIAO, as well as several biophysical characterizations which allow us to learn from the real phenomenon taking place. We found that the aggregation/oligomerization process is active in the sera and showed that it happens very fast. The RIAO can form fibers and react with anti-hIAPP and anti-amyloid oligomers antibodies. Our results opens the epistemic horizon and reveal real differences between the four groups (Controls vs obesity, T1DM or T2DM) accelerating the process of understanding and discovering novel and more efficient prevention, diagnostic, transmission and therapeutic pathways.
Collapse
Affiliation(s)
- Myriam M Altamirano-Bustamante
- Unidad de Investigación en Enfermedades Metabólicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico.
| | | | - Mateo Larralde-Laborde
- Unidad de Investigación en Enfermedades Metabólicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | | | - Edgar Leyva-García
- Unidad de Investigación en Enfermedades Metabólicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Eulalia Garrido-Magaña
- UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Gerardo Rojas
- UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | | | - Cristina Revilla-Monsalve
- Unidad de Investigación en Enfermedades Metabólicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Perla Altamirano
- Unidad de Investigación en Enfermedades Metabólicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | | |
Collapse
|
23
|
Sinézia C, Lima LMTR. Heterotropic Modulation of Amylin Fibrillation by Small Molecules: Implications for Formulative Designs. Protein J 2019; 39:10-20. [DOI: 10.1007/s10930-019-09877-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Metal binding to the amyloid-β peptides in the presence of biomembranes: potential mechanisms of cell toxicity. J Biol Inorg Chem 2019; 24:1189-1196. [PMID: 31562546 DOI: 10.1007/s00775-019-01723-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/10/2019] [Indexed: 12/18/2022]
Abstract
The amyloid-β (Aβ) peptides are key molecules in Alzheimer's disease (AD) pathology. They interact with cellular membranes, and can bind metal ions outside the membrane. Certain oligomeric Aβ aggregates are known to induce membrane perturbations and the structure of these oligomers-and their membrane-perturbing effects-can be modulated by metal ion binding. If the bound metal ions are redox active, as e.g., Cu and Fe ions are, they will generate harmful reactive oxygen species (ROS) just outside the membrane surface. Thus, the membrane damage incurred by toxic Aβ oligomers is likely aggravated when redox-active metal ions are present. The combined interactions between Aβ oligomers, metal ions, and biomembranes may be responsible for at least some of the neuronal death in AD patients.
Collapse
|
25
|
Keshavarzi F, Golsheh S. IRS1- rs10498210 G/A and CCR5-59029 A/G polymorphisms in patients with type 2 diabetes in Kurdistan. Mol Genet Genomic Med 2019; 7:e631. [PMID: 30884193 PMCID: PMC6503169 DOI: 10.1002/mgg3.631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/23/2018] [Accepted: 02/01/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The insulin receptor substrate 1 (IRS1) is a critical factor in the signaling pathway for insulin, and mutations in this gene have been reported, which contribute to the ability to develop type 2 diabetes. The polymorphisms in the promoter region of C-C motif chemokine receptor5 (CCR5) are also being studied as candidates for susceptibility to develop type 2 diabetes. The aim of the current study was to determine the relationship between IRS1 and CCR5 polymorphisms with type 2 diabetes in the Kurdistan population. METHODS Genomic DNA was isolated from the blood by salt extraction method and the polymorphisms were examined using Restriction Fragment Length Polymorphism (RFLP) method. RESULTS The results of current study indicated that the frequency of AA genotype in type 2 diabetic patients in both CCR5 (OR = 2.9, p = 0.04) and IRS1 (OR = 3.3, p = 0.036) were significantly more than controls. CONCLUSION According to the results of this study, the presence of AA genotype in both CCR5 and IRS1 is associated with type 2 diabetes. There was no significant association between AG or GG genotypes with type 2 diabetes.
Collapse
Affiliation(s)
| | - Shadi Golsheh
- Department of BiologyKurdistan Science and Research BranchIslamic Azad UniversitySanandajIran
| |
Collapse
|
26
|
Tian H, Wang ZY. Zinc Chelator Inhibits Zinc-Induced Islet Amyloid Polypeptide Deposition and Apoptosis in INS-1 Cells. Biol Trace Elem Res 2019; 189:201-208. [PMID: 30027367 DOI: 10.1007/s12011-018-1444-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/12/2018] [Indexed: 12/16/2022]
Abstract
Amyloid deposition and beta cell apoptosis are characteristic pathological features of type 2 diabetes mellitus (DM). Islet amyloid polypeptide (IAPP) is the most abundant component of amyloid deposition. Monomeric IAPP does not form amyloid deposition, but the fibrous IAPP may aggregate and form amyloid deposits. Previous studies have shown that zinc is closely related to IAPP deposition through crosslink with monomeric IAPP into fibrous aggregates. In this study, we aimed to investigate whether chelating zinc could inhibit zinc-induced amyloid deposits and apoptosis of islet beta cell. N, N, N', N'-Tetrakis (2-pyridylmethyl) ethylenediamine (TPEN) is a specific chelator of zinc, with membrane permeability. It could effectively reduce the concentration of intracellular zinc. So, we used TPEN to treat hIAPP-transfected INS-1 cells. By MTT assay, the concentration (1 μM) and incubation time (12 h) of TPEN without affecting cell viability were determined. The results showed that TPEN reduced zinc-induced IAPP deposition in the culture system. Furthermore, we analyzed the effect of zinc and TPEN on the apoptosis and insulin level. The results showed that TPEN could reverse zinc-induced INS-1 cell apoptosis and insulin secretion. And the anti-apoptosis effects of TPEN is related to extracellular regulated protein kinases (ERK)/c-jun N-terminal kinase (JNK) signaling pathway. The present data indicated that chelating zinc could inhibit zinc-induced amyloid deposition and beta cell apoptosis. Thus, maintaining zinc homeostasis in islet beta cell might become a useful strategy for DM therapy.
Collapse
Affiliation(s)
- He Tian
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of education, China Medical University, Shenyang, 110122, People's Republic of China
- Department of Histology and Embryology, Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Zhan-You Wang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of education, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
27
|
Azadirachtin inhibits amyloid formation, disaggregates pre-formed fibrils and protects pancreatic β-cells from human islet amyloid polypeptide/amylin-induced cytotoxicity. Biochem J 2019; 476:889-907. [DOI: 10.1042/bcj20180820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/18/2019] [Accepted: 02/27/2019] [Indexed: 12/16/2022]
Abstract
Abstract
The human islet amyloid polypeptide (hIAPP) or amylin is the major constituent of amyloidogenic aggregates found in pancreatic islets of type 2 diabetic patients that have been associated with β-cell dysfunction and/or death associated with type 2 diabetes mellitus (T2DM). Therefore, developing and/or identifying inhibitors of hIAPP aggregation pathway and/or compound that can mediate disaggregation of preformed aggregates holds promise as a medical intervention for T2DM management. In the current study, the anti-amyloidogenic potential of Azadirachtin (AZD)—a secondary metabolite isolated from traditional medicinal plant Neem (Azadirachta indica)—was investigated by using a combination of biophysical and cellular assays. Our results indicate that AZD supplementation not only inhibits hIAPP aggregation but also disaggregates pre-existing hIAPP fibrils by forming amorphous aggregates that are non-toxic to pancreatic β-cells. Furthermore, AZD supplementation in pancreatic β-cells (INS-1E) resulted in inhibition of oxidative stress; along with restoration of the DNA damage, lipid peroxidation and the associated membrane damage, endoplasmic reticulum stress and mitochondrial membrane potential. AZD treatment also restored glucose-stimulated insulin secretion from pancreatic islets exposed to hIAPP. All-atom molecular dynamics simulation studies on full-length hIAPP pentamer with AZD suggested that AZD interacted with four possible binding sites in the amyloidogenic region of hIAPP. In summary, our results suggest AZD to be a promising candidate for combating T2DM and related amyloidogenic disorders.
Collapse
|
28
|
Alghrably M, Czaban I, Jaremko Ł, Jaremko M. Interaction of amylin species with transition metals and membranes. J Inorg Biochem 2019; 191:69-76. [DOI: 10.1016/j.jinorgbio.2018.11.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/30/2018] [Accepted: 11/04/2018] [Indexed: 02/06/2023]
|
29
|
Influence of methionine–ruthenium complex on the fibril formation of human islet amyloid polypeptide. J Biol Inorg Chem 2019; 24:179-189. [DOI: 10.1007/s00775-019-01637-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/14/2019] [Indexed: 02/07/2023]
|
30
|
Stevenson MJ, Uyeda KS, Harder NHO, Heffern MC. Metal-dependent hormone function: the emerging interdisciplinary field of metalloendocrinology. Metallomics 2019; 11:85-110. [PMID: 30270362 PMCID: PMC10249669 DOI: 10.1039/c8mt00221e] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
For over 100 years, there has been an incredible amount of knowledge amassed concerning hormones in the endocrine system and their central role in human health. Hormones represent a diverse group of biomolecules that are released by glands, communicate signals to their target tissue, and are regulated by feedback loops to maintain organism health. Many disease states, such as diabetes and reproductive disorders, stem from misregulation or dysfunction of hormones. Increasing research is illuminating the intricate roles of metal ions in the endocrine system where they may act advantageously in concert with hormones or deleteriously catalyze hormone-associated disease states. As the critical role of metal ions in the endocrine system becomes more apparent, it is increasingly important to untangle the complex mechanisms underlying the connections between inorganic biochemistry and hormone function to understand and control endocrinological phenomena. This tutorial review harmonizes the interdisciplinary fields of endocrinology and inorganic chemistry in the newly-termed field of "metalloendocrinology". We describe examples linking metals to both normal and aberrant hormone function with a focus on highlighting insight to molecular mechanisms. Hormone activities related to both essential metal micronutrients, such as copper, iron, zinc, and calcium, and disruptive nonessential metals, such as lead and cadmium are discussed.
Collapse
Affiliation(s)
- Michael J Stevenson
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
31
|
Golsheh S, Keshavarzi F. Genetic variants linked to T2DM risk in Kurdish populations. Diabetes Metab Syndr Obes 2019; 12:431-437. [PMID: 31114273 PMCID: PMC6497875 DOI: 10.2147/dmso.s189170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 03/05/2019] [Indexed: 02/06/2023] Open
Abstract
Background: The polymorphisms of the C-C chemokine receptor type 5 (CCR5) and the insulin receptor substrate 1 (IRS1) have been studied as candidates for the susceptibility to develop type 2 diabetes mellitus (T2DM). CCR5 is a chemokine receptor, and the polymorphisms in the promoter region of this receptor are being studied as candidates for the susceptibility to develop T2DM. Also, IRS1 is a critical factor in the signaling pathway for insulin, and mutations in this gene have been reported, which contribute to the ability to develop T2DM. The aim of the current study was to determine the relationship between CCR5 (59029A/G) and IRS1 (rs10498210) polymorphisms with T2DM in Sanandajian patients. Methods: Genomic DNA was isolated from 200 healthy individuals and 220 Kurdish T2DM patients by salt extraction method and the polymorphisms were examined by restriction fragment length polymorphism (RFLP) method and then the results were analyzed using Chi-square test. Results: The frequency of AA genotype in 220 Kurdish patients for both genes CCR5 (OR=1.9, P=0.02) and IRS1 (OR [95% CI]=2.62, P=0.02) were significantly more than controls. There was no significant association between AG or GG genotypes in with T2DM. Conclusion: The presence of AA homozygote alleles in both loci of IRS1 (rs10498210) and CCR5 (59029A/G) genes increased the risk of T2DM.
Collapse
Affiliation(s)
- Shadi Golsheh
- Department of Biology, Kurdistan Science and Research Branch, Islamic Azad University, Sanandaj, Iran
| | - Fatemeh Keshavarzi
- Department of Biology, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
- Correspondence: Fatemeh KeshavarziSanandaj Branch, Islamic Azad University, Pasdaran Avenue, Sanandaj, IranTel +98 918 370 4918Fax +98 873 328 8677Email
| |
Collapse
|
32
|
Lee YH, Lin Y, Cox SJ, Kinoshita M, Sahoo BR, Ivanova M, Ramamoorthy A. Zinc boosts EGCG's hIAPP amyloid Inhibition both in solution and membrane. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1867:529-536. [PMID: 30468883 DOI: 10.1016/j.bbapap.2018.11.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/14/2018] [Accepted: 11/17/2018] [Indexed: 12/22/2022]
Abstract
Amyloid aggregation of human islet amyloid polypeptide (hIAPP) is linked to insulin-producing islet cell death in type II diabetes. Previous studies have shown that zinc (Zn(II)) and insulin, co-secreted with hIAPP, have an inhibition effect on hIAPP aggregation. Lipid membranes have also been shown to significantly influence the aggregation kinetics of hIAPP. An increasing number of studies report the importance of developing small molecule inhibitors to suppress the hIAPP's aggregation and subsequent toxicity. The ability of epigallocatechin-gallate (EGCG) to inhibit aggregation of a variety of amyloid peptide/proteins initiated numerous studies as well as the development of derivative compounds to potentially treat amyloid diseases. In this study, a combination of Thioflavin-T fluorescence kinetics, transmission electron microscopy, isothermal titration calorimetery, circular dicrosim and nucelar magnetic resonance experiments were used to demonstrate a significant enhancement in EGCG's efficiency when complexed with Zn(II). We demonstrate that the Zn-EGCG complex is able to significantly suppress hIAPP's amyloid aggregation both in presence and absence of lipid membrane. Circular dichroism experiments indicate the formation and stabilization of a helical structure of hIAPP in presence of the EGCG:Zn(II) complex. Our results also reveal the ability of EGCG or EGCG:Zn(II) to efficiently suppress hIAPP's cellular toxicity. We believe that the reported results could be useful to develop strategies to trap hIAPP intermediates for further biophysical and structural studies, and also to devise approaches to abolish amyloid aggregation and cellular toxicity.
Collapse
Affiliation(s)
- Young-Ho Lee
- Institute for Protein research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan; Protein Structure Research Group, Division of Bioconvergence Analysis, Korea Basic Science Institute, Chungcheongbuk-do 28119, South Korea
| | - Yuxi Lin
- Department of Chemistry, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul 04310, South Korea
| | - Sarah J Cox
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Misaki Kinoshita
- Institute for Protein research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| | - Bikash R Sahoo
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Magdalena Ivanova
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA.
| |
Collapse
|
33
|
Atrián-Blasco E, Gonzalez P, Santoro A, Alies B, Faller P, Hureau C. Cu and Zn coordination to amyloid peptides: From fascinating chemistry to debated pathological relevance. Coord Chem Rev 2018; 375:38-55. [PMID: 30262932 DOI: 10.1016/j.ccr.2018.04.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Several diseases share misfolding of different peptides and proteins as a key feature for their development. This is the case of important neurodegenerative diseases such as Alzheimer's and Parkinson's diseases and type II diabetes mellitus. Even more, metal ions such as copper and zinc might play an important role upon interaction with amyloidogenic peptides and proteins, which could impact their aggregation and toxicity abilities. In this review, the different coordination modes proposed for copper and zinc with amyloid-β, α-synuclein and IAPP will be reviewed as well as their impact on the aggregation, and ROS production in the case of copper. In addition, a special focus will be given to the mutations that affect metal binding and lead to familial cases of the diseases. Different modifications of the peptides that have been observed in vivo and could be relevant for the coordination of metal ions are also described.
Collapse
Affiliation(s)
- Elena Atrián-Blasco
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4, France
- University of Toulouse, UPS, INPT, 31077 Toulouse Cedex 4, France
| | - Paulina Gonzalez
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR7177), Université de Strasbourg, 4 rue B. Pascal, 67081 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| | - Alice Santoro
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR7177), Université de Strasbourg, 4 rue B. Pascal, 67081 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| | - Bruno Alies
- Université de Bordeaux, ChemBioPharm INSERM U1212 CNRS UMR 5320, Bordeaux, France
| | - Peter Faller
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR7177), Université de Strasbourg, 4 rue B. Pascal, 67081 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4, France
- University of Toulouse, UPS, INPT, 31077 Toulouse Cedex 4, France
| |
Collapse
|
34
|
Hoffmann AR, Saravanan MS, Lequin O, Killian JA, Khemtemourian L. A single mutation on the human amyloid polypeptide modulates fibril growth and affects the mechanism of amyloid-induced membrane damage. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1783-1792. [DOI: 10.1016/j.bbamem.2018.02.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/30/2022]
|
35
|
Sasahara K. Membrane-mediated amyloid deposition of human islet amyloid polypeptide. Biophys Rev 2018; 10:453-462. [PMID: 29204886 PMCID: PMC5899711 DOI: 10.1007/s12551-017-0351-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/14/2017] [Indexed: 01/01/2023] Open
Abstract
Amyloid deposition of human islet amyloid polypeptide (hIAPP) within the islet of Langerhans is closely associated with type II diabetes mellitus. Accumulating evidence indicates that the membrane-mediated aggregation and subsequent deposition of hIAPP are linked to the dysfunction and death of insulin-producing pancreatic β-cells, but the molecular process of hIAPP deposition is poorly understood. In this review, I focus on recent in vitro studies utilizing model membranes to observe the membrane-mediated aggregation/deposition of hIAPP. Membrane surfaces can serve as templates for both hIAPP adsorption and aggregation. Using high-sensitivity surface analyzing/imaging techniques that can characterize the processes of hIAPP aggregation and deposition at the membrane surface, these studies provide valuable insights into the mechanism of membrane damage caused by amyloid deposition of the peptide.
Collapse
Affiliation(s)
- Kenji Sasahara
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
36
|
Ge X, Kakinen A, Gurzov EN, Yang W, Pang L, Pilkington EH, Govindan-Nedumpully P, Chen P, Separovic F, Davis TP, Ke PC, Ding F. Zinc-coordination and C-peptide complexation: a potential mechanism for the endogenous inhibition of IAPP aggregation. Chem Commun (Camb) 2018; 53:9394-9397. [PMID: 28745731 DOI: 10.1039/c7cc04291d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aggregation of the highly amyloidogenic IAPP is endogenously inhibited inside beta-cell granules at millimolar concentrations. Combining in vitro experiments and computer simulations, we demonstrated that the stabilization of IAPP upon the formation of zinc-coordinated ion molecular complex with C-peptide might be important for the endogenous inhibition of IAPP aggregation.
Collapse
Affiliation(s)
- Xinwei Ge
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Naito A, Matsumori N, Ramamoorthy A. Dynamic membrane interactions of antibacterial and antifungal biomolecules, and amyloid peptides, revealed by solid-state NMR spectroscopy. Biochim Biophys Acta Gen Subj 2018; 1862:307-323. [PMID: 28599848 PMCID: PMC6384124 DOI: 10.1016/j.bbagen.2017.06.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/28/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022]
Abstract
A variety of biomolecules acting on the cell membrane folds into a biologically active structure in the membrane environment. It is, therefore, important to determine the structures and dynamics of such biomolecules in a membrane environment. While several biophysical techniques are used to obtain low-resolution information, solid-state NMR spectroscopy is one of the most powerful means for determining the structure and dynamics of membrane bound biomolecules such as antibacterial biomolecules and amyloidogenic proteins; unlike X-ray crystallography and solution NMR spectroscopy, applications of solid-state NMR spectroscopy are not limited by non-crystalline, non-soluble nature or molecular size of membrane-associated biomolecules. This review article focuses on the applications of solid-state NMR techniques to study a few selected antibacterial and amyloid peptides. Solid-state NMR studies revealing the membrane inserted bent α-helical structure associated with the hemolytic activity of bee venom melittin and the chemical shift oscillation analysis used to determine the transmembrane structure (with α-helix and 310-helix in the N- and C-termini, respectively) of antibiotic peptide alamethicin are discussed in detail. Oligomerization of an amyloidogenic islet amyloid polypeptide (IAPP, or also known as amylin) resulting from its aggregation in a membrane environment, molecular interactions of the antifungal natural product amphotericin B with ergosterol in lipid bilayers, and the mechanism of lipid raft formation by sphingomyelin studied using solid state NMR methods are also discussed in this review article. This article is part of a Special Issue entitled "Biophysical Exploration of Dynamical Ordering of Biomolecular Systems" edited by Dr. Koichi Kato.
Collapse
Affiliation(s)
- Akira Naito
- Graduate School of Engineering, Yokohama National University, Yokohama 240-8501, Japan.
| | - Nobuaki Matsumori
- Department of Chemistry, Graduate School of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Ayyalusamy Ramamoorthy
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109-1055, USA; Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA
| |
Collapse
|
38
|
D’Urso L, Condorelli M, Puglisi O, Tempra C, Lolicato F, Compagnini G, La Rosa C. Detection and characterization at nM concentration of oligomers formed by hIAPP, Aβ(1–40) and their equimolar mixture using SERS and MD simulations. Phys Chem Chem Phys 2018; 20:20588-20596. [DOI: 10.1039/c7cp08552d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We report a structural investigation on IAPP, Aβ(1–40) and their equimolar mixture at nM concentration using SERS spectroscopy and molecular dynamic simulations.
Collapse
Affiliation(s)
- Luisa D’Urso
- Department of Chemical Sciences
- Viale A. Doria 6-95125 Catania
- Italy
| | | | - Orazio Puglisi
- Department of Chemical Sciences
- Viale A. Doria 6-95125 Catania
- Italy
| | - Carmelo Tempra
- Department of Chemical Sciences
- Viale A. Doria 6-95125 Catania
- Italy
| | - Fabio Lolicato
- Department of Physics
- University of Helsinki
- FI-00014 Helsinki
- Finland
- Laboratory of Physics
| | | | - Carmelo La Rosa
- Department of Chemical Sciences
- Viale A. Doria 6-95125 Catania
- Italy
| |
Collapse
|
39
|
Magrì A, Pietropaolo A, Tabbì G, La Mendola D, Rizzarelli E. From Peptide Fragments to Whole Protein: Copper(II) Load and Coordination Features of IAPP. Chemistry 2017; 23:17898-17902. [PMID: 29111583 DOI: 10.1002/chem.201704910] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Indexed: 12/28/2022]
Abstract
The copper-binding features of rat islet amyloid polypeptide (r-IAPP) are herein disclosed through the determination of the stability constants and spectroscopic properties of its copper complex species. To mimic the metal binding sites of the human IAPP (h-IAPP), a soluble, single-point mutated variant of r-IAPP, having a histidine residue in place of Arg18, was synthesized, that is, r-IAPP(1-37; R18H). The peptide IAPP(1-8) was also characterized to have deeper insight into the N-terminus copper(II)-binding features of r-IAPP as well as of its mutated form. A combined experimental (thermodynamic and spectroscopic) and computational approach allowed us to assess the metal loading and the coordination features of the whole IAPP. At physiological pH, the N-terminal amino group is the Cu2+ main binding site both of entire r-IAPP and of its mutated form that mimics h-IAPP. The histidine residue present in this mutated polypeptide accounts for the second Cu2+ binding. We can speculate that the copper driven toxicity of h-IAPP in comparison to that of r-IAPP can be attributed to the different metal loading and the presence of a second metal anchoring site, the His18 , whose role is usually invoked in the process of h-IAPP aggregation.
Collapse
Affiliation(s)
- Antonio Magrì
- Istituto di Biostrutture e Bioimmagini-CNR, Via P. Gaifami 18, 95126, Catania, Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università "Magna Graecia" di Catanzaro, Campus Universitario, Viale Europa, 88100, Catanzaro, Italy
| | - Giovanni Tabbì
- Istituto di Biostrutture e Bioimmagini-CNR, Via P. Gaifami 18, 95126, Catania, Italy
| | - Diego La Mendola
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Enrico Rizzarelli
- Istituto di Biostrutture e Bioimmagini-CNR, Via P. Gaifami 18, 95126, Catania, Italy.,Dipartimento di Scienze Chimiche, Università di Catania, Viale A. Doria, 5, 95125, Catania, Italy
| |
Collapse
|
40
|
Wineman-Fisher V, Miller Y. Insight into a New Binding Site of Zinc Ions in Fibrillar Amylin. ACS Chem Neurosci 2017; 8:2078-2087. [PMID: 28692245 DOI: 10.1021/acschemneuro.7b00221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Amylin peptides are secreted together with insulin and zinc ions from pancreatic β-cells. Under unknown conditions, the amylin peptides aggregate to produce oligomers and fibrils, and in some cases Zn2+ ions can bind to amylin peptides to form Zn2+-aggregate complexes. Consequently, these aggregates lead to the death of the β-cells and a decrease in insulin, which is one of the symptoms of type-2 diabetes (T2D). Therefore, it is crucial to investigate the binding sites of the Zn2+ ions in fibrillary amylin. It was previously found by in vitro and simulation studies that Zn2+ ion binds to two or four His residues in the turn domain of fibrillary amylin. In the current study, we present a new Zn2+ binding site in the N-terminus of fibrillary amylin with three different coordination modes. Our simulations showed that Zn2+ ions bind to polymorphic amylin fibrils with a preference to bind to four Cys residues rather than two Cys residues of two neighboring amylin monomers. The new binding site leads to conformational changes, increases the number of polymorphic states, and demonstrates the existence of competition between various binding sites. Our study provides insight into the molecular mechanisms through which Zn2+ ions that play a critical role in amylin aggregation can bind to amylin and promote amylin aggregation in T2D.
Collapse
Affiliation(s)
- Vered Wineman-Fisher
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Be’er Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science
and Technology, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Be’er Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science
and Technology, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
| |
Collapse
|
41
|
Zn(II) - pramlintide: Stability, binding sites and unexpected aggregation. J Inorg Biochem 2017; 174:150-155. [DOI: 10.1016/j.jinorgbio.2017.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/31/2017] [Accepted: 06/20/2017] [Indexed: 11/23/2022]
|
42
|
Du W, Gong G, Wang W, Xu J. Regulation of the aggregation behavior of human islet amyloid polypeptide fragment by titanocene complexes. J Biol Inorg Chem 2017; 22:1065-1074. [DOI: 10.1007/s00775-017-1484-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023]
|
43
|
Su L, Lu C, Yan P, Zhang N, Cai S, Zhang G, Zhou X, Bin Li. The effect of aluminum ion on the aggregation of human islet amyloid polypeptide (11-28). Acta Biochim Biophys Sin (Shanghai) 2017; 49:355-360. [PMID: 28338927 DOI: 10.1093/abbs/gmx015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Indexed: 01/20/2023] Open
Abstract
Metal ions play a critical role in human islet amyloid polypeptide (hIAPP) aggregation, which is believed to be closely associated with β-cell death in type II diabetes. In this work, the effect of Al3+ on the aggregation of hIAPP (11-28) was studied by several different experimental approaches. Atomic force microscopy measurements showed that Al3+ could remarkably inhibit hIAPP(11-28) fibrillogenesis, while Zn2+ had a slight promotion effect on peptide aggregation, which was also confirmed by Thioflavin T fluorescence observation. Furthermore, X-ray photoelectron spectroscopy measurement indicated that Al ions might form chemical bonds with neighboring atoms and destroy the secondary structures of the protein. Our studies could deepen the understanding of the role of metal ions in the aggregation of amyloid peptides.
Collapse
Affiliation(s)
- Lanlan Su
- School of Science, Ningbo University, Ningbo 315211, China
| | - Cheng Lu
- School of Science, Ningbo University, Ningbo 315211, China
| | - Peng Yan
- School of Science, Ningbo University, Ningbo 315211, China
| | - Nan Zhang
- School of Science, Ningbo University, Ningbo 315211, China
| | - Sheng Cai
- Ningbo Institute of Material Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Gongjun Zhang
- Ningbo Institute of Material Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Xingfei Zhou
- School of Science, Ningbo University, Ningbo 315211, China
| | - Bin Li
- Laboratory of Physical Biology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| |
Collapse
|
44
|
Wang ST, Lin Y, Todorova N, Xu Y, Mazo M, Rana S, Leonardo V, Amdursky N, Spicer CD, Alexander BD, Edwards AA, Matthews SJ, Yarovsky I, Stevens MM. Facet-Dependent Interactions of Islet Amyloid Polypeptide with Gold Nanoparticles: Implications for Fibril Formation and Peptide-Induced Lipid Membrane Disruption. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2017; 29:1550-1560. [PMID: 28260837 PMCID: PMC5333186 DOI: 10.1021/acs.chemmater.6b04144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/25/2017] [Indexed: 05/03/2023]
Abstract
A comprehensive understanding of the mechanisms of interaction between proteins or peptides and nanomaterials is crucial for the development of nanomaterial-based diagnostics and therapeutics. In this work, we systematically explored the interactions between citrate-capped gold nanoparticles (AuNPs) and islet amyloid polypeptide (IAPP), a 37-amino acid peptide hormone co-secreted with insulin from the pancreatic islet. We utilized diffusion-ordered spectroscopy, isothermal titration calorimetry, localized surface plasmon resonance spectroscopy, gel electrophoresis, atomic force microscopy, transmission electron microscopy (TEM), and molecular dynamics (MD) simulations to systematically elucidate the underlying mechanism of the IAPP-AuNP interactions. Because of the presence of a metal-binding sequence motif in the hydrophilic peptide domain, IAPP strongly interacts with the Au surface in both the monomeric and fibrillar states. Circular dichroism showed that AuNPs triggered the IAPP conformational transition from random coil to ordered structures (α-helix and β-sheet), and TEM imaging suggested the acceleration of IAPP fibrillation in the presence of AuNPs. MD simulations revealed that the IAPP-AuNP interactions were initiated by the N-terminal domain (IAPP residues 1-19), which subsequently induced a facet-dependent conformational change in IAPP. On a Au(111) surface, IAPP was unfolded and adsorbed directly onto the Au surface, while for the Au(100) surface, it interacted predominantly with the citrate adlayer and retained some helical conformation. The observed affinity of AuNPs for IAPP was further applied to reduce the level of peptide-induced lipid membrane disruption.
Collapse
Affiliation(s)
- Shih-Ting Wang
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Yiyang Lin
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Nevena Todorova
- School
of Engineering, RMIT University, GPO Box 2476, Melbourne, Victoria 3001, Australia
| | - Yingqi Xu
- Department
of Life Sciences, Imperial College London, London SW7 2AZ, U.K.
| | - Manuel Mazo
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Subinoy Rana
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Vincent Leonardo
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Nadav Amdursky
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Christopher D. Spicer
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Bruce D. Alexander
- Department
of Pharmaceutical, Chemical and Environmental Science, University of Greenwich, Central Avenue, Chatham, Kent ME4 4TB, U.K.
| | - Alison A. Edwards
- Medway School
of Pharmacy, Universities of Kent and Greenwich
at Medway, Central Avenue, Chatham, Kent ME4 4TB, U.K.
| | - Steve J. Matthews
- Department
of Life Sciences, Imperial College London, London SW7 2AZ, U.K.
| | - Irene Yarovsky
- School
of Engineering, RMIT University, GPO Box 2476, Melbourne, Victoria 3001, Australia
| | - Molly M. Stevens
- Department
of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department
of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
- E-mail:
| |
Collapse
|
45
|
Ratha BN, Ghosh A, Brender JR, Gayen N, Ilyas H, Neeraja C, Das KP, Mandal AK, Bhunia A. Inhibition of Insulin Amyloid Fibrillation by a Novel Amphipathic Heptapeptide: MECHANISTIC DETAILS STUDIED BY SPECTROSCOPY IN COMBINATION WITH MICROSCOPY. J Biol Chem 2016; 291:23545-23556. [PMID: 27679488 PMCID: PMC5095409 DOI: 10.1074/jbc.m116.742460] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/24/2016] [Indexed: 02/02/2023] Open
Abstract
The aggregation of insulin into amyloid fibers has been a limiting factor in the development of fast acting insulin analogues, creating a demand for excipients that limit aggregation. Despite the potential demand, inhibitors specifically targeting insulin have been few in number. Here we report a non-toxic and serum stable-designed heptapeptide, KR7 (KPWWPRR-NH2), that differs significantly from the primarily hydrophobic sequences that have been previously used to interfere with insulin amyloid fibrillation. Thioflavin T fluorescence assays, circular dichroism spectroscopy, and one-dimensional proton NMR experiments suggest KR7 primarily targets the fiber elongation step with little effect on the early oligomerization steps in the lag time period. From confocal fluorescence and atomic force microscopy experiments, the net result appears to be the arrest of aggregation in an early, non-fibrillar aggregation stage. This mechanism is noticeably different from previous peptide-based inhibitors, which have primarily shifted the lag time with little effect on later stages of aggregation. As insulin is an important model system for understanding protein aggregation, the new peptide may be an important tool for understanding peptide-based inhibition of amyloid formation.
Collapse
Affiliation(s)
| | | | - Jeffrey R Brender
- Radiation Biology Branch, National Institutes of Health, Bethesda, Maryland 20814
| | - Nilanjan Gayen
- Department of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | | | - Chilukoti Neeraja
- TIFR Centre for Interdisciplinary Sciences (TCIS), Narsingi, Hyderabad 500075, India, and
| | - Kali P Das
- Department of Chemistry, 93/1 APC Road, Bose Institute, Kolkata 700009, India
| | - Atin K Mandal
- Department of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | | |
Collapse
|
46
|
Challenges in studying the structures of metal-amyloid oligomers related to type 2 diabetes, Parkinson's disease, and Alzheimer's disease. Coord Chem Rev 2016. [DOI: 10.1016/j.ccr.2016.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
47
|
Sánchez-López C, Cortés-Mejía R, Miotto MC, Binolfi A, Fernández CO, Del Campo JM, Quintanar L. Copper Coordination Features of Human Islet Amyloid Polypeptide: The Type 2 Diabetes Peptide. Inorg Chem 2016; 55:10727-10740. [PMID: 27704849 DOI: 10.1021/acs.inorgchem.6b01963] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human islet amyloid polypeptide (hIAPP) is the major component of amyloid deposits found in pancreatic β-cells of patients with type 2 diabetes (T2D). Copper ions have an inhibitory effect on the amyloid aggregation of hIAPP, and they may play a role in the etiology of T2D. However, deeper knowledge of the structural details of the copper-hIAPP interaction is required to understand the molecular mechanisms involved. Here, we performed a spectroscopic study of Cu(II) binding to hIAPP and several variants, using electron paramagnetic resonance (EPR), nuclear magnetic resonance (NMR), electronic absorption, and circular dichroism (CD) in the UV-vis region in combination with Born-Oppenheimer molecular dynamics (BOMD) and density functional theory geometry optimizations. We find that Cu(II) binds to the imidazole N1 of His18, the deprotonated amides of Ser19 and Ser20, and an oxygen-based ligand provided by Ser20, either via its hydroxyl group or its backbone carbonyl, while Asn22 might also play a role as an axial ligand. Ser20 plays a crucial role in stabilizing Cu(II) coordination toward the C-terminal, providing a potential link between the S20G mutation associated with early onset of T2D, its impact in Cu binding properties, and hIAPP amyloid aggregation. Our study defines the nature of the coordination environment in the Cu(II)-hIAPP complex, revealing that the amino acid residues involved in metal ion binding are also key residues for the formation of β-sheet structures and amyloid fibrils. Cu(II) binding to hIAPP may lead to the coexistence of more than one coordination mode, which in turn could favor different sets of Cu-induced conformational ensembles. Cu-induced hIAPP conformers would display a higher energetic barrier to form amyloid fibrils, hence explaining the inhibitory effect of Cu ions in hIAPP aggregation. Overall, this study provides further structural insights into the bioinorganic chemistry of T2D.
Collapse
Affiliation(s)
- Carolina Sánchez-López
- Departamento de Química, Centro de Investigación y de Estudios Avanzados (Cinvestav) , Mexico City, Mexico
| | - Rodrigo Cortés-Mejía
- Departamento de Física y Química Teórica, Facultad de Química, Universidad Nacional Autónoma de México (UNAM) , Mexico City, Mexico
| | - Marco C Miotto
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC) and Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario , Ocampo y Esmeralda, S2002LRK Rosario, Argentina
| | - Andres Binolfi
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC) and Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario , Ocampo y Esmeralda, S2002LRK Rosario, Argentina
| | - Claudio O Fernández
- Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC) and Instituto de Investigaciones para el Descubrimiento de Fármacos de Rosario (IIDEFAR, UNR-CONICET), Universidad Nacional de Rosario , Ocampo y Esmeralda, S2002LRK Rosario, Argentina
| | - Jorge M Del Campo
- Departamento de Física y Química Teórica, Facultad de Química, Universidad Nacional Autónoma de México (UNAM) , Mexico City, Mexico
| | - Liliana Quintanar
- Departamento de Química, Centro de Investigación y de Estudios Avanzados (Cinvestav) , Mexico City, Mexico
| |
Collapse
|
48
|
Erthal LCS, Marques AF, Almeida FCL, Melo GLM, Carvalho CM, Palmieri LC, Cabral KMS, Fontes GN, Lima LMTR. Regulation of the assembly and amyloid aggregation of murine amylin by zinc. Biophys Chem 2016; 218:58-70. [PMID: 27693831 DOI: 10.1016/j.bpc.2016.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 09/10/2016] [Accepted: 09/17/2016] [Indexed: 11/17/2022]
Abstract
The secretory granule of the pancreatic β-cells is a zinc-rich environment copopulated with the hormones amylin and insulin. The human amylin is shown to interact with zinc ions with major contribution from the single histidine residue, which is absent in amylin from other species such as cat, rhesus and rodents. We report here the interaction of murine amylin with zinc ions in vitro. The self-assembly of murine amylin is tightly regulated by zinc and pH. Ion mobility mass spectrometry revealed zinc interaction with monomers and oligomers. Nuclear magnetic resonance confirms the binding of zinc to murine amylin. The aggregation process of murine amylin into amyloid fibrils is accelerated by zinc. Collectively these data suggest a general role of zinc in the modulation of amylin variants oligomerization and amyloid fibril formation.
Collapse
Affiliation(s)
- Luiza C S Erthal
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil
| | - Adriana F Marques
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil
| | - Fábio C L Almeida
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil
| | - Gustavo L M Melo
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil
| | - Camila M Carvalho
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil
| | - Leonardo C Palmieri
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil
| | - Katia M S Cabral
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil
| | - Giselle N Fontes
- Laboratory for Macromolecules (LAMAC-DIMAV), Brazilian National Institute of Metrology, Quality and Technology - INMETRO, Av. N. Sa. das Graças, 50 - Xerém, Duque de Caxias-RJ, 25250-020 Rio de Janeiro, Brazil
| | - Luís Maurício T R Lima
- School of Pharmacy, Federal University of Rio de Janeiro - UFRJ, CCS, Bss24, Ilha do Fundão, 21941-590 Rio de Janeiro, Brazil; Laboratory for Macromolecules (LAMAC-DIMAV), Brazilian National Institute of Metrology, Quality and Technology - INMETRO, Av. N. Sa. das Graças, 50 - Xerém, Duque de Caxias-RJ, 25250-020 Rio de Janeiro, Brazil; National Institute of Science and Technology for Structural Biology and Bioimaging (INBEB-INCT), Federal University of Rio de Janeiro, Rio de Janeiro 21941-590, Brazil.
| |
Collapse
|
49
|
Magrì A, La Mendola D, Nicoletti VG, Pappalardo G, Rizzarelli E. New Insight in Copper-Ion Binding to Human Islet Amyloid: The Contribution of Metal-Complex Speciation To Reveal the Polypeptide Toxicity. Chemistry 2016; 22:13287-300. [PMID: 27493030 DOI: 10.1002/chem.201602816] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Indexed: 01/05/2023]
Abstract
Type-2 diabetes (T2D) is considered to be a potential threat on a global level. Recently, T2D has been listed as a misfolding disease, such as Alzheimer's and Parkinson's diseases. Human islet amyloid polypeptide (hIAPP) is a molecule cosecreted in pancreatic β cells and represents the main constituent of an aggregated amyloid found in individuals affected by T2D. The trace-element serum level is significantly influenced during the development of diabetes. In particular, the dys-homeostasis of Cu(2+) ions may adversely affect the course of the disease. Conflicting results have been reported on the protective role played by complex species formed by Cu(2+) ions with hIAPP or its peptide fragments in vitro. The histidine (His) residue at position 18 represents the main binding site for the metal ion, but contrasting results have been reported on other residues involved in metal-ion coordination, in particular those toward the N or C terminus. Sequences that encompass regions 17-29 and 14-22 were used to discriminate between the two models of the hIAPP coordination mode. Due to poor solubility in water, poly(ethylene glycol) (PEG) derivatives were synthesized. A peptide fragment that encompasses the 17-29 region of rat amylin (rIAPP) in which the arginine residue at position 18 was substituted by a histidine residue was also obtained to assess that the PEG moiety does not alter the peptide secondary structure. The complex species formed by Cu(2+) ions with Ac-PEG-hIAPP(17-29)-NH2 , Ac-rIAPP(17-29)R18H-NH2 , and Ac-PEG-hIAPP(14-22)-NH2 were studied by using potentiometric titrations coupled with spectroscopic methods (UV/Vis, circular dichroism, and EPR). The combined thermodynamic and spectroscopic approach allowed us to demonstrate that hIAPP is able to bind Cu(2+) ions starting from the His18 imidazole nitrogen atom toward the N-terminus domain. The stability constants of copper(II) complexes with Ac-PEG-hIAPP(14-22)-NH2 were used to simulate the different experimental conditions under which aggregate formation and oxidative stress of hIAPP has been reported. Speciation unveils: 1) the protective role played by increased amounts of Cu(2+) ions on the hIAPP fibrillary aggregation, 2) the effect of adventitious trace amounts of Cu(2+) ions present in phosphate-buffered saline (PBS), and 3) a reducing fluorogenic probe on H2 O2 production attributed to the polypeptide alone.
Collapse
Affiliation(s)
- Antonio Magrì
- Consiglio Nazionale delle Ricerche, Istituto di Biostrutture e Bioimmagini, Via P. Gaifami 18, 95126, Catania, Italy.
| | - Diego La Mendola
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano, 6, 56126, Pisa, Italy.
| | - Vincenzo Giuseppe Nicoletti
- Dipartimento di Scienze Biomediche e Biotecnologiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Pappalardo
- Consiglio Nazionale delle Ricerche, Istituto di Biostrutture e Bioimmagini, Via P. Gaifami 18, 95126, Catania, Italy
| | - Enrico Rizzarelli
- Consiglio Nazionale delle Ricerche, Istituto di Biostrutture e Bioimmagini, Via P. Gaifami 18, 95126, Catania, Italy.,Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
50
|
Amphiphilic xanthones as a potent chemical entity of anti-mycobacterial agents with membrane-targeting properties. Eur J Med Chem 2016; 123:684-703. [PMID: 27517813 DOI: 10.1016/j.ejmech.2016.07.068] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022]
Abstract
Tuberculosis (TB) remains a deadly disease and infects one-third of the world's population. Given the low success rates encountered in clinical development, there is an urgent need to identify structurally novel antimicrobials for tuberculosis. The present report details the anti-mycobacterial activities, structure-activity relationships (SARs) and mechanism of action of amphiphilic xanthone derivatives. The xanthones exhibited potent MIC, rapid time-kill and no cross-resistance with the current anti-TB drugs. Evidence is presented that these compounds disrupted the inner membrane and led to ATP depletion. Amphiphilic xanthone derivatives exhibited superior metabolic stability, low cytotoxicity and low activity against the common cytochrome P450. Compound 5 was selected for an in vivo pharmacokinetic study. Its bioavailability at an oral dose of 2 mg/kg was 15%. The xanthones thuse provide valuable insight for the development of a new class of membrane targeting antimycobacterial agents that may assist in overcoming the limitations of the current TB medications.
Collapse
|