1
|
Schmaltz L, Barakat E, Fleurot R, Uzbekov R, Reynaud K, Laffont L, Tsikis G, Mérour I, Mermillod P, Saint-Dizier M. Phosphatidylserine on sperm head interact with Annexin A5 on oviduct luminal cilia to form a sperm reservoir in pigs. Eur J Cell Biol 2025; 104:151471. [PMID: 39700614 DOI: 10.1016/j.ejcb.2024.151471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/05/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024] Open
Abstract
After insemination, a subpopulation of sperm reaches the oviducts and binds to isthmic epithelial cells to form a "sperm reservoir". Our objective was to explore the role of annexin A5 (ANXA5), a protein that binds with high affinity to phosphatidylserine (PS), in the formation of the sperm reservoir in pigs. Phosphatidylserine was detected on the head of approximately 10 % of boar sperm at ejaculation. Porcine ANXA5 was immunodetected with a strong signal on luminal cilia in the isthmus and in derived isthmic epithelial spheroids (IES). Exogenous PS between 0.01 and 0.1 µg/mL and recombinant porcine ANXA5 (rpANXA5) above 0.1 µg/mL inhibited sperm binding to IES without reducing sperm motility. Pre-incubation of sperm, but not IES, with rpANXA5 inhibited sperm binding to IES. Under capacitating conditions, the proportion of live sperm with head PS exposure and the ability of sperm to bind to rpANXA5 and IES cilia increased within 30 min. Conversely, the acrosome reaction decreased the ability of sperm to bind rpANXA5 and prevented sperm binding to IES. In conclusion, sperm membrane remodelling during capacitation enhanced head PS exposure in motile sperm, resulting in increased interaction with ciliary ANXA5 on isthmic epithelial spheroids. These findings support a role for PS-ANXA5 interaction in the formation of the sperm reservoir in mammalian females.
Collapse
Affiliation(s)
- Lorraine Schmaltz
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France
| | - Elie Barakat
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France
| | - Renaud Fleurot
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France
| | - Rustem Uzbekov
- Laboratory of Cell Biology and Electron Microscopy, University of Tours, Tours, France
| | - Karine Reynaud
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France
| | - Ludivine Laffont
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France
| | - Guillaume Tsikis
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France
| | | | - Pascal Mermillod
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France
| | - Marie Saint-Dizier
- INRAE, CNRS, University of Tours, Physiologie de la Reproduction et des comportements, Center INRAE Val-de-Loire, Nouzilly, France.
| |
Collapse
|
2
|
Sitton J, Pickett D, Rodriguez A, Kurouski D. Lipids determine the toxicity of human islet polypeptide aggregates in vivo. J Biol Chem 2025; 301:108029. [PMID: 39615682 PMCID: PMC11728924 DOI: 10.1016/j.jbc.2024.108029] [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: 08/01/2024] [Revised: 11/16/2024] [Accepted: 11/25/2024] [Indexed: 12/22/2024] Open
Abstract
The onset and progression of type 2 diabetes is linked to the accumulation and aggregation of human islet amyloid polypeptide (hIAPP) in the pancreas. Amyloid oligomers and fibrils formed as a result of such aggregation exert high cytotoxicity. Although some pieces of evidence suggest that lipids could alter the rate of hIAPP aggregation, the effect of lipids on the aggregation properties of this peptide remains unclear. In this study, we investigate the effect of sphingophospholipid and anionic and zwitterionic phospholipids with different lengths of fatty acids on the aggregation of hIAPP. We found that anionic lipids drastically accelerate peptide aggregation, whereas this effect was substantially weaker for sphingophospholipid and zwitterionic phospholipid. Biophysical analysis revealed that the presence of lipids resulted in substantial differences in morphology and secondary structure of hIAPP fibrils compared to the protein aggregates grown in the lipid-free environment. We also found that zwitterionic phospholipids drastically increased cytotoxicity of hIAPP aggregates, whereas this effect was less evident for sphingophospholipid and anionic phospholipid. Our results showed that drastic differences in lipid-determined cytotoxicity of hIAPP aggregates were linked to molecular mechanisms of autophagy, exocytosis, and unfolded protein response. These findings suggest that molecular candidates that could disrupt protein-lipid interactions would allow for deceleration of the onset and progression of type 2 diabetes.
Collapse
Affiliation(s)
- Jadon Sitton
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States
| | - Davis Pickett
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States
| | - Axell Rodriguez
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States; Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States.
| |
Collapse
|
3
|
Kolobova E, Petrushanko I, Mitkevich V, Makarov AA, Grigorova IL. β-Amyloids and Immune Responses Associated with Alzheimer's Disease. Cells 2024; 13:1624. [PMID: 39404388 PMCID: PMC11475064 DOI: 10.3390/cells13191624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is associated with the accumulation of β-amyloids (Aβs) and the formation of Aβ plaques in the brain. Various structural forms and isoforms of Aβs that have variable propensities for oligomerization and toxicity and may differentially affect the development of AD have been identified. In addition, there is evidence that β-amyloids are engaged in complex interactions with the innate and adaptive immune systems, both of which may also play a role in the regulation of AD onset and progression. In this review, we discuss what is currently known about the intricate interplay between β-amyloids and the immune response to Aβs with a more in-depth focus on the possible roles of B cells in the pathogenesis of AD.
Collapse
Affiliation(s)
- Elizaveta Kolobova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Irina Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Vladimir Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Irina L Grigorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| |
Collapse
|
4
|
Zhaliazka K, Kurouski D. Elucidation of molecular mechanisms by which amyloid β 1-42 fibrils exert cell toxicity. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159510. [PMID: 38759921 DOI: 10.1016/j.bbalip.2024.159510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
Abrupt aggregation of amyloid β1-42 (Aβ1-42) peptide in the frontal lobe is the expected underlying cause of Alzheimer's disease (AD). β-Sheet-rich oligomers and fibrils formed by Aβ1-42 exert high cell toxicity. A growing body of evidence indicates that lipids can uniquely alter the secondary structure and toxicity of Aβ1-42 aggregates. At the same time, underlying molecular mechanisms that determine this difference in toxicity of amyloid aggregates remain unclear. Using a set of molecular and biophysical assays to determine the molecular mechanism by which Aβ1-42 aggregates formed in the presence of cholesterol, cardiolipin, and phosphatidylcholine exert cell toxicity. Our findings demonstrate that rat neuronal cells exposed to Aβ1-42 fibrils formed in the presence of lipids with different chemical structure exert drastically different magnitude and dynamic of unfolded protein response (UPR) in the endoplasmic reticulum (ER) and mitochondria (MT). We found that the opposite dynamics of UPR in MT and ER in the cells exposed to Aβ1-42: cardiolipin fibrils and Aβ1-42 aggregates formed in a lipid-free environment. We also found that Aβ1-42: phosphatidylcholine fibrils upregulated ER UPR simultaneously downregulating the UPR response of MT, whereas Aβ1-42: cholesterol fibrils suppressed the UPR response of ER and upregulated UPR response of MT. We also observed progressively increasing ROS production that damages mitochondrial membranes and other cell organelles, ultimately leading to cell death.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
5
|
Yin X, Zhou H, Cao T, Yang X, Meng F, Dai X, Wang Y, Li S, Zhai W, Yang Z, Chen N, Zhou R. Rational Design of Dual-Functionalized Gd@C 82 Nanoparticles to Relieve Neuronal Cytotoxicity in Alzheimer's Disease via Inhibition of Aβ Aggregation. ACS NANO 2024; 18:15416-15431. [PMID: 38840269 DOI: 10.1021/acsnano.3c08823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
The accumulation of amyloid-β (Aβ) peptides is a major hallmark of Alzheimer's disease (AD) and plays a crucial role in its pathogenesis. Particularly, the structured oligomeric species rich in β-sheet formations were implicated in neuronal organelle damage. Addressing this formidable challenge requires identifying candidates capable of inhibiting peptide aggregation or disaggregating preformed oligomers for effective antiaggregation-based AD therapy. Here, we present a dual-functional nanoinhibitor meticulously designed to target the aggregation driving force and amyloid fibril spatial structure. Leveraging the exceptional structural stability and facile tailoring capability of endohedral metallofullerene Gd@C82, we introduce desired hydrogen-binding sites and charged groups, which are abundant on its surface for specific designs. Impressively, these designs endow the resultant functionalized-Gd@C82 nanoparticles (f-Gd@C82 NPs) with high capability of redirecting peptide self-assembly toward disordered, off-pathway species, obstructing the early growth of protofibrils, and disaggregating the preformed well-ordered protofibrils or even mature Aβ fibrils. This results in considerable alleviation of Aβ peptide-induced neuronal cytotoxicity, rescuing neuronal death and synaptic loss in primary neuron models. Notably, these modifications significantly improved the dispersibility of f-Gd@C82 NPs, thus substantially enhancing its bioavailability. Moreover, f-Gd@C82 NPs demonstrate excellent cytocompatibility with various cell lines and possess the ability to penetrate the blood-brain barrier in mice. Large-scale molecular dynamics simulations illuminate the inhibition and disaggregation mechanisms. Our design successfully overcomes the limitations of other nanocandidates, which often overly rely on hydrophobic interactions or photothermal conversion properties, and offers a viable direction for developing anti-AD agents through the inhibition and even reversal of Aβ aggregation.
Collapse
Affiliation(s)
- Xiuhua Yin
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Hong Zhou
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Tiantian Cao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
- Suzhou Institute of Trade and Commerce, Suzhou 215009, China
| | - Xiner Yang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Fei Meng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Xing Dai
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Yifan Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Sijie Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Wangsong Zhai
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Zaixing Yang
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Ning Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| |
Collapse
|
6
|
Qiang W, Kengewerere MK, Kenyaga JM. Modulation of Lipid Dynamics in the β-Amyloid Aggregates Induced Membrane Fragmentation. J Phys Chem B 2024; 128:5667-5675. [PMID: 38836448 DOI: 10.1021/acs.jpcb.4c02119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Nonspecific membrane disruption is considered a plausible mechanism for the cytotoxicity induced by β-amyloid (Aβ) aggregates. In scenarios of high local Aβ concentrations, a two-step membrane fragmentation model has been proposed. Initially, membrane-embedded Aβ oligomeric aggregates form, followed by membrane fragmentation. However, the key molecular-level interactions between Aβ oligomeric aggregates and lipids that drive the second-stage membrane fragmentation remain unclear. This study monitors the time-dependent changes in lipid dynamics and water accessibility of model liposomes during Aβ-induced membrane fragmentation. Our results indicate that lipid dynamics on the nanosecond to microsecond time scale undergo rapid acceleration upon initial incubation with membrane-incorporated Aβ oligomeric aggregates, followed by a slow deceleration process. Concurrently, lipid headgroups become less accessible to water. Both observations suggest a carpet-like mechanism of membrane disruption for the Aβ-induced membrane fragmentation process.
Collapse
Affiliation(s)
- Wei Qiang
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Maurine K Kengewerere
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - June M Kenyaga
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| |
Collapse
|
7
|
Ali A, Dou T, Holman AP, Hung A, Osborne L, Pickett D, Rodriguez A, Zhaliazka K, Kurouski D. The influence of zwitterionic and anionic phospholipids on protein aggregation. Biophys Chem 2024; 306:107174. [PMID: 38211368 DOI: 10.1016/j.bpc.2024.107174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
The progressive aggregation of misfolded proteins is the underlying molecular cause of numerous pathologies including Parkinson's disease and injection and transthyretin amyloidosis. A growing body of evidence indicates that protein deposits detected in organs and tissues of patients diagnosed with such pathologies contain fragments of lipid membranes. In vitro experiments also showed that lipid membranes could strongly change the aggregation rate of amyloidogenic proteins, as well as alter the secondary structure and toxicity of oligomers and fibrils formed in their presence. In this review, the effect of large unilamellar vesicles (LUVs) composed of zwitterionic and anionic phospholipids on the aggregation rate of insulin, lysozyme, transthyretin (TTR) and α- synuclein (α-syn) will be discussed. The manuscript will also critically review the most recent findings on the lipid-induced changes in the secondary structure of protein oligomers and fibrils, as well as reveal the extent to which lipids could alter the toxicity of protein aggregates formed in their presence.
Collapse
Affiliation(s)
- Abid Ali
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Tianyi Dou
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Aidan P Holman
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Entomology, Texas A&M University, College Station, TX 77843, United States
| | - Andrew Hung
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Luke Osborne
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Davis Pickett
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Axell Rodriguez
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
8
|
Serravalle S, Pisano M, Sciacca MFM, Salamone N, Sicali L, Mazzara G, Costa L, La Rosa C. Critical micellar concentration determination of pure phospholipids and lipid-raft and their mixtures with cholesterol. Proteins 2024. [PMID: 38234101 DOI: 10.1002/prot.26669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/01/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024]
Abstract
Phospholipids in biological membranes establish a chemical equilibrium between free phospholipids in the aqueous phase (CMC) and self-assembled phospholipids in vesicles, keeping the CMC constant. The CMC is different for each phospholipid, depends on the amount of cholesterol, and, according to the lipid-chaperone hypothesis, controls the interaction between free phospholipids and amyloidogenic proteins (such as amylin, amyloid-β, and α-synuclein, all of which are, respectively, associated with a different proteinopathy), which governs the formation of a toxic complex between free lipids and proteins that leads to membrane destruction. Here, we provide quantitative measurements of CMCs and bilayer stability of pure phospholipids, lipid rafts, and their mixture with cholesterol by fluorescence methods (using pyrene as a probe) and light scattering techniques (resonance Rayleigh scattering and fixed-angle light scattering) performed on LUVs, as well as AFM to measure LUV dimensions. Also, we test the lipid-chaperone hypothesis on human IAPP interacting with different mixture of POPC cholesterol. Stated the importance of CMC in membrane stability and protein aggregation processes, these results could be a starting point for the development of a quantitative kinetic model for the lipid chaperone hypothesis.
Collapse
Affiliation(s)
- Sofia Serravalle
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Martina Pisano
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Michele F M Sciacca
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Catania, Italy
| | - Nancy Salamone
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Luciano Sicali
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Giuseppe Mazzara
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Luca Costa
- Centre de Biologie Structurale, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Carmelo La Rosa
- Department of Chemical Sciences, University of Catania, Catania, Italy
| |
Collapse
|
9
|
Regmi D, Shen F, Stanic A, Islam M, Du D. Effect of phospholipid liposomes on prion fragment (106-128) amyloid formation. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184199. [PMID: 37454869 DOI: 10.1016/j.bbamem.2023.184199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/08/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Misfolding and aggregation of cellular prion protein (PrPc) is a major molecular process involved in the pathogenesis of prion diseases. Here, we studied the aggregation properties of a prion fragment peptide PrP(106-128). The results show that the peptide aggregates in a concentration-dependent manner in an aqueous solution and that the aggregation is sensitive to pH and the preformed amyloid seeds. Furthermore, we show that the zwitterionic POPC liposomes moderately inhibit the aggregation of PrP(106-128), whereas POPC/cholesterol (8:2) vesicles facilitate peptide aggregation likely due to the increase of the lipid packing order and membrane rigidity in the presence of cholesterol. In addition, anionic lipid vesicles of POPG and POPG/cholesterol above a certain concentration accelerate the aggregation of the peptide remarkably. The strong electrostatic interactions between the N-terminal region of the peptide and POPG may constrain the conformational plasticity of the peptide, preventing insertion of the peptide into the inner side of the membrane and thus promoting fibrillation on the membrane surface. The results suggest that the charge properties of the membrane, the composition of the liposomes, and the rigidity of lipid packing are critical in determining peptide adsorption on the membrane surface and the efficiency of the membrane in catalyzing peptide oligomeric nucleation and amyloid formation. The peptide could be used as an improved model molecule to investigate the mechanistic role of the crucial regions of PrP in aggregation in a membrane-rich environment and to screen effective inhibitors to block key interactions between these regions and membranes for preventing PrP aggregation.
Collapse
Affiliation(s)
- Deepika Regmi
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Fengyun Shen
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Aleksander Stanic
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Majedul Islam
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Deguo Du
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA.
| |
Collapse
|
10
|
Wu R, Wang Z, Jia Z, Li C, Wang J, Liu L, Dong M. Identification of hybrid amyloid strains assembled from amyloid- βand human islet amyloid polypeptide. NANOTECHNOLOGY 2023; 34:505101. [PMID: 37625382 DOI: 10.1088/1361-6528/acf3ee] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023]
Abstract
Cross-fibrillation of amyloid-β(Aβ) peptides and human islet amyloid polypeptides (hIAPP) has revealed a close correlation between Alzheimer's disease and type 2 diabetes (T2D). Importantly, different amyloid strains are likely to lead to the clinical pathological heterogeneity of degenerative diseases due to toxicity. However, given the complicated cross-interactions between different amyloid peptides, it is still challenging to identify the polymorphism of the hybrid amyloid strains and reveal mechanistic insights into aggregation, but highly anticipated due to their significance. In this study, we investigated the cross-fibrillation of Aβpeptides and different hIAPP species (monomers, oligomers, and fibrils) using combined experimental and simulation approaches. Cross-seeding and propagation of different amyloid peptides monitored by experimental techniques proved that the three species of hIAPP aggregates have successively enhanced Aβfibrillation, especially for hIAPP fibrils. Moreover, the polymorphism of these morphologically similar hybrid amyloid strains could be distinguished by testing their mechanical properties using quantitative nanomechanical mapping, where the assemblies of Aβ-hIAPP fibrils exhibited the high Young's modulus. Furthermore, the enhanced internal molecular interactions andβ-sheet structural transformation were proved by exploring the conformational ensembles of Aβ-hIAPP heterodimer and Aβ-hIAPP decamer using molecular dynamic simulations. Our findings pave the way for identifying different hybrid amyloid strains by quantitative nanomechanical mapping and molecular dynamic simulations, which is important not only for the precise classification of neurodegenerative disease subtypes but also for future molecular diagnosis and therapeutic treatment of multiple interrelated degenerative diseases.
Collapse
Affiliation(s)
- Rongrong Wu
- Institute for Advanced Materials, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Zengkai Wang
- Institute for Advanced Materials, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Zili Jia
- Institute for Advanced Materials, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Chenglong Li
- Institute for Advanced Materials, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Jie Wang
- Institute for Advanced Materials, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Lei Liu
- Institute for Advanced Materials, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Mingdong Dong
- Aarhus University, Interdisciplinary Nanoscience Center (iNANO) Aarhus C DK-8000, Denmark
| |
Collapse
|
11
|
Recoulat Angelini AA, Incicco JJ, Melian NA, González-Flecha FL. Susceptibility of Cu(I) transport ATPases to sodium dodecyl sulfate. Relevance of the composition of the micellar phase. Arch Biochem Biophys 2023; 745:109704. [PMID: 37527700 DOI: 10.1016/j.abb.2023.109704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/11/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
Sodium dodecyl sulfate (SDS) is a well-known protein denaturing agent. A less known property of this detergent is that it can activate or inactivate some enzymes at sub-denaturing concentrations. In this work we explore the effect of SDS on the ATPase activity of a hyper-thermophilic and a mesophilic Cu(I) ATPases reconstituted in mixed micelles of phospholipids and a non-denaturing detergent. An iterative procedure was used to evaluate the partition of SDS between the aqueous and the micellar phases, allowing to determine the composition of micelles prepared from phospholipid/detergent mixtures. The incubation of enzymes with SDS in the presence of different amounts of phospholipids reveals that higher SDS concentrations are required to obtain the same degree of inactivation when the initial concentration of phospholipids is increased. Remarkably, we found that, if represented as a function of the mole fraction of SDS in the micelle, the degree of inactivation obtained at different amounts of amphiphiles converges to a single inactivation curve. To interpret this result, we propose a simple model involving active and inactive enzyme molecules in equilibrium. This model allowed us to estimate the Gibbs free energy change for the inactivation process and its derivative with respect to the mole fraction of SDS in the micellar phase, the latter being a measure of the susceptibility of the enzyme to SDS. Our results showed that the inactivation free energy changes are similar for both proteins. Conversely, susceptibility to SDS is significantly lower for the hyperthermophilic ATPase, suggesting an inverse relation between thermophilicity and susceptibility to SDS.
Collapse
Affiliation(s)
- Alvaro A Recoulat Angelini
- Universidad de Buenos Aires - CONICET, Laboratorio de Biofísica Molecular. Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - J Jeremías Incicco
- Universidad de Buenos Aires - CONICET, Laboratorio de Biofísica Molecular. Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - Noelia A Melian
- Universidad de Buenos Aires - CONICET, Laboratorio de Biofísica Molecular. Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - F Luis González-Flecha
- Universidad de Buenos Aires - CONICET, Laboratorio de Biofísica Molecular. Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina.
| |
Collapse
|
12
|
Kumari A, Saha D, Bhattacharya J, Aswal VK, Moulick RG. Studying the structural organization of non-membranous protein hemoglobin in a lipid environment after reconstitution. Int J Biol Macromol 2023:125212. [PMID: 37302629 DOI: 10.1016/j.ijbiomac.2023.125212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/13/2023]
Abstract
In our current work we have developed a supported 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) bilayer with embedded hemoglobin, reconstituted via detergent-mediated method. Microscopic studies revealed that the hemoglobin molecules could be visualized without any labelling agents. The reconstituted proteins assemble themselves as supramolecular structures to adapt to lipid bilayer environment. The nonionic detergent, n-octyl-β-d-glucoside (NOG) used for insertion of hemoglobin played an important role in formation of these structures. When concentrations of lipid, protein and detergent were raised by four folds, we observed phase separation by protein molecules within bilayer via protein-protein assembly. This phase separation process exhibited extremely slow kinetics to form large stable domains with correlation times in the order of minutes. Confocal Z-scanning images showed that these supramolecular structures generated membrane deformities. UV-Vis, Fluorescence and Circular Dichroism (CD) measurement indicated minor structural change to expose the hydrophobic regions of the protein to adjust the hydrophobic stress of the lipid environment whilst Small Angle Neutron Scattering (SANS) results indicated that the hemoglobin molecules retained their overall tetrameric form in the system. In conclusion, we state that this investigation allowed us to closely inspect some rare but noteworthy phenomena like the formation of supramolecular structures, large domain formation and membrane deformation etc.
Collapse
Affiliation(s)
- Akanksha Kumari
- Amity Institute of Biotechnology, Amity University Haryana, 122413, India
| | - Debasish Saha
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | | | - V K Aswal
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | | |
Collapse
|
13
|
Zhaliazka K, Serada V, Matveyenka M, Rizevsky S, Kurouski D. Protein-to-lipid ratio uniquely changes the rate of lysozyme aggregation but does not significantly alter toxicity of mature protein aggregates. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159305. [PMID: 36907244 PMCID: PMC10405292 DOI: 10.1016/j.bbalip.2023.159305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/09/2023] [Accepted: 03/01/2023] [Indexed: 03/13/2023]
Abstract
Irreversible aggregation of misfolded proteins is the underlying molecular cause of numerous pathologies, including diabetes type 2, Alzheimer's, and Parkinson's diseases. Such an abrupt protein aggregation results in the formation of small oligomers that can propagate into amyloid fibrils. A growing body of evidence suggests that protein aggregation can be uniquely altered by lipids. However, the role of the protein-to-lipid (P:L) ratio on the rate of protein aggregation, as well as the structure and toxicity of corresponding protein aggregates remains poorly understood. In this study, we investigate the role of the P:L ratio of five different phospho- and sphingolipids on the rate of lysozyme aggregation. We observed significantly different rates of lysozyme aggregation at 1:1, 1:5, and 1:10 P:L ratios of all analyzed lipids except phosphatidylcholine (PC). However, we found that at those P:L ratios, structurally and morphologically similar fibrils were formed. As a result, for all studies of lipids except PC, mature lysozyme aggregates exerted insignificantly different cell toxicity. These results demonstrate that the P:L ratio directly determines the rate of protein aggregation, however, has very little if any effect on the secondary structure of mature lysozyme aggregates. Furthermore, our results point to the lack of a direct relationship between the rate of protein aggregation, secondary structure, and toxicity of mature fibrils.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Valeryia Serada
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Stanislav Rizevsky
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biotechnology, Binh Duong University, Thu Dau Mot 820000, Viet Nam
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
14
|
Sebastiao M, Babych M, Quittot N, Kumar K, Arnold AA, Marcotte I, Bourgault S. Development of a novel fluorescence assay for studying lipid bilayer perturbation induced by amyloidogenic peptides using cell plasma membrane vesicles. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184118. [PMID: 36621762 DOI: 10.1016/j.bbamem.2022.184118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/22/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
Numerous pathophysiological conditions are associated with the misfolding and aggregation of proteins into insoluble amyloid fibrils. The mechanisms by which this process leads to cellular dysfunction remain elusive, though several hypotheses point toward the perturbation of the cell plasma membrane by pre-fibrillar intermediates and/or amyloid growth. However, current models to study membrane perturbations are largely limited to synthetic lipid vesicles and most of experimental approaches cannot be transposed to complex cell-derived plasma membrane systems. Herein, vesicles originating from the plasma membrane of erythrocytes and β-pancreatic cells were used to study the perturbations induced by an amyloidogenic peptide, the islet amyloid polypeptide (IAPP). These biologically relevant lipid vesicles displayed a characteristic clustering in the presence of the amyloidogenic peptide, which was able to rupture membranes. By exploiting Förster resonance energy transfer (FRET), a rapid, simple, and potentially high-throughput assay to detect membrane perturbations of intact mammalian cell plasma membrane vesicles was implemented. The FRET kinetics of membrane perturbations closely correlated with the kinetics of thioflavin-T fluorescence associated with amyloid formation. This novel kinetics assay expands the toolbox available to study amyloid-associated membrane damage, bridging the gap between synthetic lipid vesicles and living cells.
Collapse
Affiliation(s)
- Mathew Sebastiao
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Quebec, QC, Canada
| | - Margaryta Babych
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Quebec, QC, Canada
| | - Noé Quittot
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Quebec, QC, Canada
| | - Kiran Kumar
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Quebec, QC, Canada
| | - Alexandre A Arnold
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Quebec, QC, Canada
| | - Isabelle Marcotte
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Quebec, QC, Canada.
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Quebec, QC, Canada.
| |
Collapse
|
15
|
Rando C, Grasso G, Sarkar D, Sciacca MFM, Cucci LM, Cosentino A, Forte G, Pannuzzo M, Satriano C, Bhunia A, La Rosa C. GxxxG Motif Stabilize Ion-Channel like Pores through C α-H···O Interaction in Aβ (1-40). Int J Mol Sci 2023; 24:ijms24032192. [PMID: 36768518 PMCID: PMC9917128 DOI: 10.3390/ijms24032192] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Aβ (1-40) can transfer from the aqueous phase to the bilayer and thus form stable ion-channel-like pores where the protein has alpha-helical conformation. The stability of the pores is due to the presence of the GXXXG motif. It has been reported that these ion-channel-like pores are stabilized by a Cα-H···O hydrogen bond that is established between a glycine of the GXXXG sequence of an alpha-helix and another amino acid of a vicinal alpha-helix. However, conflicting data are reported in the literature. Some authors have suggested that hydrogen bonding does not have a stabilizing function. Here we synthesized pentapeptides having a GXXXG motif to explore its role in pore stability. We used molecular dynamics simulations, quantum mechanics, and experimental biophysical techniques to determine whether hydrogen bonding was formed and had a stabilizing function in ion-channel-like structures. Starting from our previous molecular dynamics data, molecular quantum mechanics simulations, and ATR data showed that a stable ion-channel-like pore formed and a band centered at 2910 cm-1 was attributed to the interaction between Gly 7 of an alpha-helix and Asp 23 of a vicinal alpha-helix.
Collapse
Affiliation(s)
- Carola Rando
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Grasso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Dibakar Sarkar
- Department of Biophysics Bose Institute, Unified Academic Campus, Bidhan Nagar, EN 80, Kolkata 700091, India
| | | | - Lorena Maria Cucci
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Alessia Cosentino
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Forte
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Cristina Satriano
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Anirban Bhunia
- Department of Biophysics Bose Institute, Unified Academic Campus, Bidhan Nagar, EN 80, Kolkata 700091, India
| | - Carmelo La Rosa
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy
- Correspondence:
| |
Collapse
|
16
|
Burrelli A, Moretti P, Gerelli Y, Ortore MG. Effects of model membranes on lysozyme amyloid aggregation. Biomol Concepts 2023; 14:bmc-2022-0034. [PMID: 37542518 DOI: 10.1515/bmc-2022-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/13/2023] [Indexed: 08/07/2023] Open
Abstract
The study of the interaction between lipid membranes and amyloidogenic peptides is a turning point for understanding the processes involving the cytotoxicity of peptides involved in neurodegenerative diseases. In this work, we perform an experimental study of model membrane-lysozyme interaction to understand how the formation of amyloid fibrils can be affected by the presence of polar and zwitterionic phospholipid molecules (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine [POPC] and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol [POPG]). The study was conducted above and below the critical micellar concentration (CMC) using dynamic light scattering (DLS), atomic force microscopy (AFM), UV-Vis spectrophotometry, and the quartz crystal microbalance (QCM). Our results show that the presence of phospholipids appears to be a factor favoring the formation of amyloid aggregates. Spectrophotometric and DLS data revealed that the quantity of β -structure increases in the presence of POPG and POPC at different concentrations. The presence of POPG and POPC increases the speed of the nucleation process, without altering the overall structures of the fibrillar final products.
Collapse
Affiliation(s)
- Annaclaudia Burrelli
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Paolo Moretti
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Yuri Gerelli
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
- CNR Institute for Complex Systems, Piazzale Aldo Moro 5, 00185 Roma, Italy
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Maria Grazia Ortore
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
17
|
Reichelderfer VT, Chaparro Sosa AF, Kaar JL, Schwartz DK. Tuning the surface charge of phospholipid bilayers inhibits insulin fibrilization. Colloids Surf B Biointerfaces 2022; 220:112904. [PMID: 36265317 PMCID: PMC10164472 DOI: 10.1016/j.colsurfb.2022.112904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/27/2022] [Accepted: 10/02/2022] [Indexed: 11/27/2022]
Abstract
The interactions between proteins and materials, in particular lipid bilayers, have been studied extensively for their relevance in diseases and for the formulation of protein-based therapeutics and vaccines. However, the precise rules by which material properties induce favorable or unfavorable structural states in biomolecules are incompletely understood, and as a result, the rational design of materials remains challenging. Here, we investigated the influence of lipid bilayers (in the form of small unilamellar vesicles) on the formation of insulin amyloid fibrils using a fibril-specific assay (thioflavin T), polyacrylamide gel electrophoresis, and circular dichroism spectroscopy. Lipid bilayers composed of equal mixtures of cationic and anionic lipids effectively inhibited fibril formation and stabilized insulin in its native conformation. However, other lipid bilayer compositions failed to inhibit fibril formation or even destabilized insulin, exacerbating fibrilization and/or non-amyloid aggregation. Our findings suggest that electrostatic interactions with lipid bilayers can play a critical role in stabilizing or destabilizing insulin, and preventing the conversion of insulin to its amyloidogenic, disease-associated state.
Collapse
Affiliation(s)
- Victoria T Reichelderfer
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
| | - Andres F Chaparro Sosa
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
| | - Joel L Kaar
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA.
| | - Daniel K Schwartz
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
18
|
Matveyenka M, Rizevsky S, Kurouski D. Amyloid aggregates exert cell toxicity causing irreversible damages in the endoplasmic reticulum. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166485. [PMID: 35840040 PMCID: PMC10424722 DOI: 10.1016/j.bbadis.2022.166485] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/08/2022] [Accepted: 07/05/2022] [Indexed: 12/31/2022]
Abstract
Amyloid oligomers and fibrils are protein aggregates that cause an onset and progression of many neurodegenerative diseases, diabetes type 2 and systemic amyloidosis. Although a growing body of evidence shows that oligomers and fibrils trigger mitochondrial dysfunction simultaneously enhancing production of reactive oxygen species, exact mechanisms by which these protein aggregates exert their toxicities remain unclear. In this study, we used advanced microscopic and spectroscopic methods to examine topography and structure of insulin aggregates grown in the lipid-free environment, as well as in the presence of major classes of phospho- and sphingolipids. We also employed a set of molecular markers to determine the extent to which insulin aggregates induce a damage of cell endoplasmic reticulum (ER), an important cell organelle used for calcium storage, protein synthesis and folding. Our results show that insulin aggregates activate the expression of Activating Transcription Factor 6 (ATF6), a transmembrane protein that is involved in unfolded protein response (UPR) of the stressed ER. At the same time, two other ER transmembrane proteins, Inositol Requiring 1 (IRE1α) and eLF2a, the product of PKR-like ER kinase (PERK), exhibited very low expression levels. Furthermore, amyloid aggregates trigger an expression of the 78-kDa glucose-regulated protein GRP78, which is also involved in the UPR. We also observed UPR-induced expression of a proapoptotic transcription factor CHOP, which, in turn, regulates expression of caspase 3 kinase and BCL2 protein family members, including the ER localized Bax. These findings show that insulin oligomers and fibrils induce UPR-associated ER stress and ultimately fatal changes in cell homeostasis.
Collapse
Affiliation(s)
- Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Stanislav Rizevsky
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biotechnology, Binh Duong University, Thu Dau Mot 820000, Viet Nam
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
19
|
Ferreira I, Rauter AP, Bandarra NM. Marine Sources of DHA-Rich Phospholipids with Anti-Alzheimer Effect. Mar Drugs 2022; 20:662. [PMID: 36354985 PMCID: PMC9695993 DOI: 10.3390/md20110662] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 07/29/2023] Open
Abstract
Alzheimer's disease (AD) is a complex and progressive disease, which affects millions of people around the world. Despite the many efforts over the years to find efficient therapeutics, there is no cure yet. Nonetheless, many compounds have been proven to decrease Alzheimer's symptoms. After a short overview of the hypotheses considered in AD drug development and the drugs approved for AD treatment, which lead to symptom release, we focus on the valorization of natural marine sources that decrease AD symptoms, particularly on docosahexaenoic acid (DHA), an important component in membrane phospholipids and the most abundant n-3 polyunsaturated fatty acids (PUFA) found in gray matter of the brain and in retina and on the DHA-containing phospholipids (DHA-PLs) present in marine sources, namely fish, krill, mollusks and in fisheries and aquaculture by-products. DHA-PLs' bioactivities are presented, namely their properties in anti-neurodegeneration, neuroinflammation, as anticancer agents, as well as their benefits to obesity and visual problems. Fisheries and aquaculture by-products are also highlighted as they have a high content of DHA and DHA-rich phospholipids, can be extracted by green methodologies and should be considered in a circular economy for a healthy sustainable future.
Collapse
Affiliation(s)
- Inês Ferreira
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Division of Aquaculture, Upgrading and Bioprospecting, Portuguese Institute of the Sea and Atmosphere, 1495-165 Lisboa, Portugal
| | - Amélia P. Rauter
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Narcisa M. Bandarra
- Division of Aquaculture, Upgrading and Bioprospecting, Portuguese Institute of the Sea and Atmosphere, 1495-165 Lisboa, Portugal
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4050-123 Porto, Portugal
| |
Collapse
|
20
|
Chakravorty A, McCalpin SD, Sahoo BR, Ramamoorthy A, Brooks CL. Free Gangliosides Can Alter Amyloid-β Aggregation. J Phys Chem Lett 2022; 13:9303-9308. [PMID: 36174129 PMCID: PMC9700483 DOI: 10.1021/acs.jpclett.2c02362] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
A recently proposed lipid-chaperone hypothesis suggests that free lipid molecules, not bound to membranes, affect the aggregation of amyloidogenic peptides such as amyloid-β (Aβ) peptides, whose aggregates are the hallmarks of Alzheimer's disease. Here, we combine experiments with all-atom molecular dynamics simulations in explicit solvent to explore the effects of neuronal ganglioside GM1, abundant in mammalian brains, on the aggregation of two principal isoforms of Aβ, Aβ40 and Aβ42. Our simulations show that free GM1 forms stable, highly water-soluble complexes with both isoforms, and nuclear magnetic resonance experiments support the formation of well-ordered, structurally compact GM1+Aβ complexes. By simulation, we also show that Aβ40 monomers display a preference for binding to GM1-containing hetero-oligomers over GM1-lacking homo-oligomers, while Aβ42 monomers have the opposite preference. These observations explain why GM1 dose-dependently inhibits Aβ40 aggregation but has no effect on Aβ42 aggregation, as assessed by thioflavin T fluorescence.
Collapse
Affiliation(s)
- Arghya Chakravorty
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, United States
| | - Samuel D. McCalpin
- Department of Chemistry, Biophysics Program, Biomedical Engineering, Macromolecular Engineering and Science, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Bikash R. Sahoo
- Department of Chemistry, Biophysics Program, Biomedical Engineering, Macromolecular Engineering and Science, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Ayyalusamy Ramamoorthy
- Department of Chemistry, Biophysics Program, Biomedical Engineering, Macromolecular Engineering and Science, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Charles L. Brooks
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, United States
- Department of Chemistry, Biophysics Program, Biomedical Engineering, Macromolecular Engineering and Science, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| |
Collapse
|
21
|
Matveyenka M, Zhaliazka K, Rizevsky S, Kurouski D. Lipids uniquely alter secondary structure and toxicity of lysozyme aggregates. FASEB J 2022; 36:e22543. [PMID: 36094052 PMCID: PMC10427241 DOI: 10.1096/fj.202200841r] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/03/2022] [Accepted: 08/29/2022] [Indexed: 08/17/2023]
Abstract
Abrupt aggregation of misfolded proteins is a hallmark of the large group of amyloid pathologies that include diabetes type 2, Alzheimer and Parkinson's diseases. Protein aggregation yields oligomers and fibrils, β-sheet-rich structures that exert cell toxicity. Microscopic examination of amyloid deposits reveals the presence of lipids membranes, which suggests that lipids can be involved in the process of pathogenic protein assembly. In this study, we show that lipids can uniquely alter the aggregation rates of lysozyme, a protein that is associated with systemic amyloidosis. Specifically, cardiolipin (CL), ceramide (CER), and sphingomyelin (SM) accelerate, phosphatidylcholine (PC) strongly inhibits, whereas phosphatidylserine (PS) has no effect on the rate of protein aggregation. Furthermore, lipids uniquely alter the secondary structure of lysozyme aggregates. Furthermore, we found that lysozyme aggregates grown in the presence of CL, CER, SM, PS, and CL:PC mixtures exert significantly lower production of reactive oxygen species and mitochondrial dysfunction compared to lysozyme:PC aggregates and lysozyme fibrils grown in the lipid-free environment. These findings suggest that a change in the lipid composition of cell membranes, which is taken place upon neurodegeneration, may trigger the formation of toxic protein species that otherwise would not be formed.
Collapse
Affiliation(s)
- Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Stanislav Rizevsky
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
- Department of Biotechnology, Binh Duong University, Thu Dau Mot, Vietnam
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
22
|
Zhaliazka K, Rizevsky S, Matveyenka M, Serada V, Kurouski D. Charge of Phospholipids Determines the Rate of Lysozyme Aggregation but Not the Structure and Toxicity of Amyloid Aggregates. J Phys Chem Lett 2022; 13:8833-8839. [PMID: 36111888 PMCID: PMC10405293 DOI: 10.1021/acs.jpclett.2c02126] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Biophysical properties of plasma membranes are determined by a chemical structure of phospholipids, including saturation of fatty acids and charge of polar heads of these molecules. Phospholipids not only determine fluidity and plasticity of membranes but also play an important role in abrupt aggregation of misfolded proteins. In this study, we investigate the role of the charge of the most abundant phospholipids in the plasma membrane on the aggregation properties of the lysozyme. We found that the charge of phospholipids determines the aggregation rate of lysozyme and the morphology of the protein aggregates. However, the secondary structure and toxicity of these protein specimens are determined by the chemical nature rather than the charge of phospholipids. These findings show that the charge of phospholipids can be a key factor that determines the stability and aggregation mechanism of amyloidogenic proteins.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Stanislav Rizevsky
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Biotechnology, Binh Duong University, Thu Dau Mot 820000, Vietnam
| | - Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Valeryia Serada
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
23
|
Wu H, Tatiyaborworntham N, Hajimohammadi M, Decker EA, Richards MP, Undeland I. Model systems for studying lipid oxidation associated with muscle foods: Methods, challenges, and prospects. Crit Rev Food Sci Nutr 2022; 64:153-171. [PMID: 35916770 DOI: 10.1080/10408398.2022.2105302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lipid oxidation is a complex process in muscle-based foods (red meat, poultry and fish) causing severe quality deterioration, e.g., off-odors, discoloration, texture defects and nutritional loss. The complexity of muscle tissue -both composition and structure- poses as a formidable challenge in directly clarifying the mechanisms of lipid oxidation in muscle-based foods. Therefore, different in vitro model systems simulating different aspects of muscle have been used to study the pathways of lipid oxidation. In this review, we discuss the principle, preparation, implementation as well as advantages and disadvantages of seven commonly-studied model systems that mimic either compositional or structural aspects of actual meat: emulsions, fatty acid micelles, liposomes, microsomes, erythrocytes, washed muscle mince, and muscle homogenates. Furthermore, we evaluate the prospects of stem cells, tissue cultures and three-dimensional printing for future model system development. Based on this reviewing of oxidation models, tailoring correct model to different study aims could be facilitated, and readers are becoming acquainted with advantages and shortcomings. In addition, insight into recent technology developments, e.g., stem cell- and tissue-cultures as well as three-dimensional printing could provide new opportunities to overcome the current bottlenecks of lipid oxidation studies in muscle.
Collapse
Affiliation(s)
- Haizhou Wu
- Department of Biology and Biological Engineering-Food and Nutrition Science, Chalmers University of Technology, Gothenburg, SE, Sweden
| | - Nantawat Tatiyaborworntham
- Food Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, Thailand
| | | | - Eric A Decker
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, USA
| | - Mark P Richards
- Department of Animal and Dairy Sciences, Meat Science and Animal Biologics Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | - Ingrid Undeland
- Department of Biology and Biological Engineering-Food and Nutrition Science, Chalmers University of Technology, Gothenburg, SE, Sweden
| |
Collapse
|
24
|
Toma I, Porfire AS, Tefas LR, Berindan-Neagoe I, Tomuță I. A Quality by Design Approach in Pharmaceutical Development of Non-Viral Vectors with a Focus on miRNA. Pharmaceutics 2022; 14:1482. [PMID: 35890377 PMCID: PMC9322860 DOI: 10.3390/pharmaceutics14071482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 07/14/2022] [Indexed: 12/10/2022] Open
Abstract
Cancer is the leading cause of death worldwide. Tumors consist of heterogeneous cell populations that have different biological properties. While conventional cancer therapy such as chemotherapy, radiotherapy, and surgery does not target cancer cells specifically, gene therapy is attracting increasing attention as an alternative capable of overcoming these limitations. With the advent of gene therapy, there is increasing interest in developing non-viral vectors for genetic material delivery in cancer therapy. Nanosystems, both organic and inorganic, are the most common non-viral vectors used in gene therapy. The most used organic vectors are polymeric and lipid-based delivery systems. These nanostructures are designed to bind and protect the genetic material, leading to high efficiency, prolonged gene expression, and low toxicity. Quality by Design (QbD) is a step-by-step approach that investigates all the factors that may affect the quality of the final product, leading to efficient pharmaceutical development. This paper aims to provide a new perspective regarding the use of the QbD approach for improving the quality of non-viral vectors for genetic material delivery and their application in cancer therapy.
Collapse
Affiliation(s)
- Ioana Toma
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| | - Alina Silvia Porfire
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| | - Lucia Ruxandra Tefas
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania;
| | - Ioan Tomuță
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| |
Collapse
|
25
|
Investigation of structure–stability correlations of reconstructed oil bodies. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Benhamou Goldfajn N, Tang H, Ding F. Substoichiometric Inhibition of Insulin against IAPP Aggregation Is Attenuated by the Incompletely Processed N-Terminus of proIAPP. ACS Chem Neurosci 2022; 13:2006-2016. [PMID: 35704461 DOI: 10.1021/acschemneuro.2c00231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Substoichiometric aggregation inhibition of human islet amyloid polypeptide (IAPP), the hallmark of type 2 diabetes impacting millions of people, is crucial for developing clinic therapies, yet it remains challenging given that many candidate inhibitors require high doses. Intriguingly, insulin, the key regulatory polypeptide on blood glucose levels that are cosynthesized, costored, and cosecreted with IAPP by pancreatic β cells, has been identified as a potent inhibitor that can suppress IAPP amyloid aggregation at substoichiometric concentrations. Here, we computationally investigated the molecular mechanisms of the substoichiometric inhibition of insulin against the aggregation of IAPP and the incompletely processed IAPP (proIAPP) using discrete molecular dynamics simulations. Our results suggest that the amyloid aggregations of both IAPP and proIAPP might be disrupted by insulin through its binding with the shared amyloidogenic core sequences. However, the N-terminus of proIAPP competed with the amyloidogenic core sequences for the insulin interactions, resulting in attenuated inhibition by insulin. Moreover, insulin preferred to bind the elongation surfaces of IAPP seeds with fibril-like structure, with a stronger affinity than that of IAPP monomers. The capping of elongation surfaces by a small amount of insulin sterically prohibited the seed growth via monomer addition, achieving the substoichiometric inhibition. Together, our computational results provided molecular insights for the substoichiometric inhibition of insulin against IAPP aggregation, also the weakened effect on proIAPP. The uncovered substoichiometric inhibition by capping the elongation of amyloid seeds or fibrils may guide the rational designs of new potent inhibitors effective at low doses.
Collapse
Affiliation(s)
- Nadav Benhamou Goldfajn
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, United States.,University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Huayuan Tang
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, United States
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, United States
| |
Collapse
|
27
|
Xie H, Rojas A, Maisuradze GG, Khelashvili G. Mechanistic Kinetic Model Reveals How Amyloidogenic Hydrophobic Patches Facilitate the Amyloid-β Fibril Elongation. ACS Chem Neurosci 2022; 13:987-1001. [PMID: 35258946 PMCID: PMC8986627 DOI: 10.1021/acschemneuro.1c00801] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Abnormal aggregation of amyloid β (Aβ) peptides into fibrils plays a critical role in the development of Alzheimer's disease. A two-stage "dock-lock" model has been proposed for the Aβ fibril elongation process. However, the mechanisms of the Aβ monomer-fibril binding process have not been elucidated with the necessary molecular-level precision, so it remains unclear how the lock phase dynamics leads to the overall in-register binding of the Aβ monomer onto the fibril. To gain mechanistic insights into this critical step during the fibril elongation process, we used molecular dynamics (MD) simulations with a physics-based coarse-grained UNited-RESidue (UNRES) force field and sampled extensively the dynamics of the lock phase process, in which a fibril-bound Aβ(9-40) peptide rearranged to establish the native docking conformation. Analysis of the MD trajectories with Markov state models was used to quantify the kinetics of the rearrangement process and the most probable pathways leading to the overall native docking conformation of the incoming peptide. These revealed a key intermediate state in which an intra-monomer hairpin is formed between the central core amyloidogenic patch 18VFFA21 and the C-terminal hydrophobic patch 34LMVG37. This hairpin structure is highly favored as a transition state during the lock phase of the fibril elongation. We propose a molecular mechanism for facilitation of the Aβ fibril elongation by amyloidogenic hydrophobic patches.
Collapse
Affiliation(s)
- Hengyi Xie
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10065, United States
| | - Ana Rojas
- Schrödinger, Inc., 1540 Broadway, 24th Floor, New York, New York 10036, United States
| | - Gia G. Maisuradze
- Baker Laboratory of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10065, United States
| |
Collapse
|
28
|
Tempra C, Scollo F, Pannuzzo M, Lolicato F, La Rosa C. A unifying framework for amyloid-mediated membrane damage: The lipid-chaperone hypothesis. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140767. [PMID: 35144022 DOI: 10.1016/j.bbapap.2022.140767] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/16/2022]
Abstract
Over the past thirty years, researchers have highlighted the role played by a class of proteins or polypeptides that forms pathogenic amyloid aggregates in vivo, including i) the amyloid Aβ peptide, which is known to form senile plaques in Alzheimer's disease; ii) α-synuclein, responsible for Lewy body formation in Parkinson's disease and iii) IAPP, which is the protein component of type 2 diabetes-associated islet amyloids. These proteins, known as intrinsically disordered proteins (IDPs), are present as highly dynamic conformational ensembles. IDPs can partially (mis) fold into (dys) functional conformations and accumulate as amyloid aggregates upon interaction with other cytosolic partners such as proteins or lipid membranes. In addition, an increasing number of reports link the toxicity of amyloid proteins to their harmful effects on membrane integrity. Still, the molecular mechanism underlying the amyloidogenic proteins transfer from the aqueous environment to the hydrocarbon core of the membrane is poorly understood. This review starts with a historical overview of the toxicity models of amyloidogenic proteins to contextualize the more recent lipid-chaperone hypothesis. Then, we report the early molecular-level events in the aggregation and ion-channel pore formation of Aβ, IAPP, and α-synuclein interacting with model membranes, emphasizing the complexity of these processes due to their different spatial-temporal resolutions. Next, we underline the need for a combined experimental and computational approach, focusing on the strengths and weaknesses of the most commonly used techniques. Finally, the last two chapters highlight the crucial role of lipid-protein complexes as molecular switches among ion-channel-like formation, detergent-like, and fibril formation mechanisms and their implication in fighting amyloidogenic diseases.
Collapse
Affiliation(s)
- Carmelo Tempra
- Institute of Organic Chemistry and Biochemistry, Prague, Czech Republic
| | - Federica Scollo
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Fabio Lolicato
- Heidelberg University Biochemistry Center, Heidelberg, Germany; Department of Physics, University of Helsinki, Helsinki, Finland.
| | - Carmelo La Rosa
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Catania, Italy.
| |
Collapse
|
29
|
Al Adem K, Shanti A, Srivastava A, Homouz D, Thomas SA, Khair M, Stefanini C, Chan V, Kim TY, Lee S. Linking Alzheimer’s Disease and Type 2 Diabetes: Characterization and Inhibition of Cytotoxic Aβ and IAPP Hetero-Aggregates. Front Mol Biosci 2022; 9:842582. [PMID: 35372522 PMCID: PMC8968156 DOI: 10.3389/fmolb.2022.842582] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
The cytotoxic self-aggregation of β-amyloid (Aβ) peptide and islet amyloid polypeptide (IAPP) is implicated in the pathogenesis of Alzheimer’s disease (AD) and Type 2 diabetes (T2D), respectively. Increasing evidence, particularly the co-deposition of Aβ and IAPP in both brain and pancreatic tissues, suggests that Aβ and IAPP cross-interaction may be responsible for a pathological link between AD and T2D. Here, we examined the nature of IAPP-Aβ40 co-aggregation and its inhibition by small molecules. In specific, we characterized the kinetic profiles, morphologies, secondary structures and toxicities of IAPP-Aβ40 hetero-assemblies and compared them to those formed by their homo-assemblies. We demonstrated that monomeric IAPP and Aβ40 form stable hetero-dimers and hetero-assemblies that further aggregate into β-sheet-rich hetero-aggregates that are toxic (cell viability <50%) to both PC-12 cells, a neuronal cell model, and RIN-m5F cells, a pancreatic cell model for β-cells. We then selected polyphenolic candidates to inhibit IAPP or Aβ40 self-aggregation and examined the inhibitory effect of the most potent candidate on IAPP-Aβ40 co-aggregation. We demonstrated that epigallocatechin gallate (EGCG) form inter-molecular hydrogen bonds with each of IAPP and Aβ40. We also showed that EGCG reduced hetero-aggregate formation and resulted in lower β-sheets content and higher unordered structures in IAPP-Aβ40-EGCG samples. Importantly, we showed that EGCG is highly effective in reducing the toxicity of IAPP-Aβ40 hetero-aggregates on both cell models, specifically at concentrations that are equivalent to or are 2.5-fold higher than the mixed peptide concentrations. To the best of our knowledge, this is the first study to report the inhibition of IAPP-Aβ40 co-aggregation by small molecules. We conclude that EGCG is a promising candidate to prevent co-aggregation and cytotoxicity of IAPP-Aβ40, which in turn, contribute to the pathological link between AD and T2D.
Collapse
Affiliation(s)
- Kenana Al Adem
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Aya Shanti
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Amit Srivastava
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dirar Homouz
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Physics, University of Houston, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - Sneha Ann Thomas
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Vincent Chan
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Tae-Yeon Kim
- Department of Civil Infrastructure and Environmental Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Sungmun Lee
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Khalifa University’s Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- *Correspondence: Sungmun Lee,
| |
Collapse
|
30
|
Mrdenovic D, Pieta IS, Nowakowski R, Kutner W, Lipkowski J, Pieta P. Amyloid β interaction with model cell membranes - What are the toxicity-defining properties of amyloid β? Int J Biol Macromol 2022; 200:520-531. [PMID: 35074328 DOI: 10.1016/j.ijbiomac.2022.01.117] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 01/26/2023]
Abstract
Disruption of the neuronal membrane by toxic amyloid β oligomers is hypothesized to be the major event associated with Alzheimer's disease's neurotoxicity. Misfolding of amyloid β is followed by aggregation via different pathways in which structurally different amyloid β oligomers can be formed. The respective toxic actions of these structurally diverse oligomers can vary significantly. Linking a particular toxic action to a structurally unique kind of amyloid β oligomers and resolving their toxicity-determining feature remains challenging because of their transient stability and heterogeneity. Moreover, the lipids that make up the membrane affect amyloid β oligomers' behavior, thus adding to the problem's complexity. The present review compares and analyzes the latest results to improve understanding of amyloid β oligomers' interaction with lipid bilayers.
Collapse
Affiliation(s)
- Dusan Mrdenovic
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland; Department of Chemistry, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1, Canada
| | - Izabela S Pieta
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Robert Nowakowski
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Wlodzimierz Kutner
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland; Faculty of Mathematics and Natural Sciences, School of Sciences, Cardinal Stefan Wyszynski University in Warsaw, Wóycickiego 1/3, 01-815 Warsaw, Poland
| | - Jacek Lipkowski
- Department of Chemistry, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1, Canada
| | - Piotr Pieta
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland.
| |
Collapse
|
31
|
Nguyen TH, Nguyen PH, Ngo ST, Derreumaux P. Effect of Cholesterol Molecules on Aβ1-42 Wild-Type and Mutants Trimers. Molecules 2022; 27:molecules27041395. [PMID: 35209177 PMCID: PMC8879133 DOI: 10.3390/molecules27041395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease displays aggregates of the amyloid-beta (Aβ) peptide in the brain, and there is increasing evidence that cholesterol may contribute to the pathogenesis of the disease. Though many experimental and theoretical studies have focused on the interactions of Aβ oligomers with membrane models containing cholesterol, an understanding of the effect of free cholesterol on small Aβ42 oligomers is not fully established. To address this question, we report on replica exchange with a solute tempering simulation of an Aβ42 trimer with cholesterol and compare it with a previous replica exchange molecular dynamics simulation. We show that the binding hot spots of cholesterol are rather complex, involving hydrophobic residues L17–F20 and L30–M35 with a non-negligible contribution of loop residues D22–K28 and N-terminus residues. We also examine the effects of cholesterol on the trimers of the disease-causing A21G and disease-protective A2T mutations by molecular dynamics simulations. We show that these two mutations moderately impact cholesterol-binding modes. In our REST2 simulations, we find that cholesterol is rarely inserted into aggregates but rather attached as dimers and trimers at the surface of Aβ42 oligomers. We propose that cholesterol acts as a glue to speed up the formation of larger aggregates; this provides a mechanistic link between cholesterol and Alzheimer’s disease.
Collapse
Affiliation(s)
- Trung Hai Nguyen
- Laboratory of Theoretical and Computational Biophysics, Ton Duc Thang University, Ho Chi Minh City, Vietnam; (T.H.N.); (S.T.N.)
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Phuong H. Nguyen
- Laboratoire de Biochimie Théorique, UPR 9080 CNRS, Université de Paris, 13 rue Pierre et Marie Curie, 75005 Paris, France;
- Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, PSL Research University, 75005 Paris, France
| | - Son Tung Ngo
- Laboratory of Theoretical and Computational Biophysics, Ton Duc Thang University, Ho Chi Minh City, Vietnam; (T.H.N.); (S.T.N.)
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Philippe Derreumaux
- Laboratoire de Biochimie Théorique, UPR 9080 CNRS, Université de Paris, 13 rue Pierre et Marie Curie, 75005 Paris, France;
- Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, PSL Research University, 75005 Paris, France
- Institut Universitaire de France (IUF), 75005 Paris, France
- Correspondence:
| |
Collapse
|
32
|
Molecular Mechanisms of Amylin Turnover, Misfolding and Toxicity in the Pancreas. Molecules 2022; 27:molecules27031021. [PMID: 35164285 PMCID: PMC8838401 DOI: 10.3390/molecules27031021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/24/2022] [Accepted: 01/29/2022] [Indexed: 12/13/2022] Open
Abstract
Amyloidosis is a common pathological event in which proteins self-assemble into misfolded soluble and insoluble molecular forms, oligomers and fibrils that are often toxic to cells. Notably, aggregation-prone human islet amyloid polypeptide (hIAPP), or amylin, is a pancreatic hormone linked to islet β-cells demise in diabetics. The unifying mechanism by which amyloid proteins, including hIAPP, aggregate and kill cells is still matter of debate. The pathology of type-2 diabetes mellitus (T2DM) is characterized by extracellular and intracellular accumulation of toxic hIAPP species, soluble oligomers and insoluble fibrils in pancreatic human islets, eventually leading to loss of β-cell mass. This review focuses on molecular, biochemical and cell-biology studies exploring molecular mechanisms of hIAPP synthesis, trafficking and degradation in the pancreas. In addition to hIAPP turnover, the dynamics and the mechanisms of IAPP–membrane interactions; hIAPP aggregation and toxicity in vitro and in situ; and the regulatory role of diabetic factors, such as lipids and cholesterol, in these processes are also discussed.
Collapse
|
33
|
Abioye RO, Okagu OD, Udenigwe CC. Inhibition of Islet Amyloid Polypeptide Fibrillation by Structurally Diverse Phenolic Compounds and Fibril Disaggregation Potential of Rutin and Quercetin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:392-402. [PMID: 34964624 DOI: 10.1021/acs.jafc.1c06918] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The influence of 12 food-derived phenolic compounds on islet amyloid polypeptide (IAPP) fibrillation was investigated. Results from thioflavin T assay demonstrated that gallic acid, caffeic acid, and rutin and its aglycone, quercetin, inhibited IAPP fibrillation at 1:0.5, 1:1, and 1:2 IAPP-phenolic molar ratios. Circular dichroism and dynamic light scattering at the 1:1 IAPP-phenolic ratio confirmed the inhibition of fibril formation. Rutin and quercetin increased the lag time by 90 and 6%, and the relative α-helix content by 63 and 48%, respectively. Gallic acid decreased the elongation rate by 30%, whereas caffeic acid decreased the maximum fluorescence intensity by 65%. Furthermore, fluorescence microscopy and transmission electron microscopy (TEM) showed IAPP fibril morphologies indicative of fibrillation reduction by the compounds. Molecular docking and TEM showed that rutin and quercetin disaggregated preformed IAPP fibrils potentially through fibrillar-monomeric equilibrium shifts. These findings demonstrate important structural features of phenolic compounds for disaggregating IAPP fibrils or inhibiting their formation.
Collapse
Affiliation(s)
- Raliat O Abioye
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Ogadimma D Okagu
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Chibuike C Udenigwe
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa K1H 8M5, Canada
| |
Collapse
|
34
|
Oliveira MC, Yusupov M, Bogaerts A, Cordeiro RM. Distribution of lipid aldehydes in phase-separated membranes: A molecular dynamics study. Arch Biochem Biophys 2022; 717:109136. [DOI: 10.1016/j.abb.2022.109136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/02/2022]
|
35
|
Ben-Zichri S, Malishev R, Oren O, Bloch DN, Taube R, Papo N, Jelinek R. Bcl-2-Homology-Only Proapoptotic Peptides Modulate β-Amyloid Aggregation and Toxicity. ACS Chem Neurosci 2021; 12:4554-4563. [PMID: 34806861 DOI: 10.1021/acschemneuro.1c00611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Aggregation of the β-Amyloid (Aβ) peptide in brain tissues is the hallmark of Alzheimer's disease (AD). While Aβ is presumed to be insidiously involved in the disease's pathophysiology, concrete mechanisms accounting for the role of Aβ in AD are yet to be deciphered. While Aβ has been primarily identified in the extracellular space, the peptide also accumulates in cellular compartments such as mitochondria and lysosomes and impairs cellular functions. Here, we show that prominent proapoptotic peptides associated with the mitochondrial outer membrane, the Bcl-2-homology-only peptides BID, PUMA, and NOXA, exert significant and divergent effects upon aggregation, cytotoxicity, and membrane interactions of Aβ42, the main Aβ homolog. Interestingly, we show that BID and PUMA accelerated aggregation of Aβ42, reduced Aβ42-induced toxicity and mitochondrial disfunction, and inhibited Aβ42-membrane interactions. In contrast, NOXA exhibited opposite effects, reducing Aβ42 fibril formation, affecting more pronounced apoptotic effects and mitochondrial disfunction, and enhancing membrane interactions of Aβ42. The effects of BID, PUMA, and NOXA upon the Aβ42 structure and toxicity may be linked to its biological properties and affect pathophysiological features of AD.
Collapse
Affiliation(s)
- Shani Ben-Zichri
- Department of Chemistry and Ilse Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ravit Malishev
- Department of Chemistry and Ilse Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ofek Oren
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Daniel N. Bloch
- Department of Chemistry and Ilse Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ran Taube
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Raz Jelinek
- Department of Chemistry and Ilse Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
36
|
Fatafta H, Kav B, Bundschuh BF, Loschwitz J, Strodel B. Disorder-to-order transition of the amyloid-β peptide upon lipid binding. Biophys Chem 2021; 280:106700. [PMID: 34784548 DOI: 10.1016/j.bpc.2021.106700] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022]
Abstract
There is mounting evidence that Alzheimer's disease progression and severity are linked to neuronal membrane damage caused by aggregates of the amyloid-β (Aβ) peptide. However, the detailed mechanism behind the membrane damage is not well understood yet. Recently, the lipid-chaperone hypothesis has been put forward, based on which the formation of complexes between Aβ and free lipids enables an easy insertion of Aβ into membranes. In order to test this hypothesis, we performed numerous all-atom molecular dynamics simulations. We studied the complex formation between individual lipids, considering both POPC and DPPC, and Aβ and examined whether the resulting complexes would be able to insert into lipid membranes. Complex formation at a one-to-one ratio was readily observed, yet with minimal effects on Aβ's characteristics. Most importantly, the peptide remains largely disordered in 1:1 complexes, and the complex does not insert into the membrane; instead, it is adsorbed to the membrane surface. The results change considerably once Aβ forms a complex with a POPC cluster composed of three lipid molecules. The hydrophobic interactions between Aβ and the lipid tails cause the peptide to fold into either a helical or a β-sheet structure. These observations provide atomic insight into the disorder-to-order transition that is needed for membrane insertion or amyloid aggregation to proceed.
Collapse
Affiliation(s)
- Hebah Fatafta
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Batuhan Kav
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Bastian F Bundschuh
- Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, Universitütstrasse 1, 40225 Düsseldorf, Germany
| | - Jennifer Loschwitz
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany; Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, Universitütstrasse 1, 40225 Düsseldorf, Germany
| | - Birgit Strodel
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany; Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, Universitütstrasse 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
37
|
Pathak BK, Dey S, Mozumder S, Sengupta J. The role of membranes in function and dysfunction of intrinsically disordered amyloidogenic proteins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:397-434. [PMID: 35034725 DOI: 10.1016/bs.apcsb.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Membrane-protein interactions play a major role in human physiology as well as in diseases pathology. Interaction of a protein with the membrane was previously thought to be dependent on well-defined three-dimensional structure of the protein. In recent decades, however, it has become evident that a large fraction of the proteome, particularly in eukaryotes, stays disordered in solution and these proteins are termed as intrinsically disordered proteins (IDPs). Also, a vast majority of human proteomes have been reported to contain substantially long disordered regions, called intrinsically disordered regions (IDRs), in addition to the structurally ordered regions. IDPs exist in an ensemble of conformations and the conformational flexibility enables IDPs to achieve functional diversity. IDPs (and IDRs) are found to be important players in cell signaling, where biological membranes act as anchors for signaling cascades. Therefore, IDPs modulate the membrane architectures, at the same time membrane composition also affects the binding of IDPs. Because of intrinsic disorders, misfolding of IDPs often leads to formation of oligomers, protofibrils and mature fibrils through progressive self-association. Accumulation of amyloid-like aggregates of some of the IDPs is a known causative agent for numerous diseases. In this chapter we highlight recent advances in understanding membrane interactions of some of the intrinsically disordered proteins involved in the pathogenesis of human diseases.
Collapse
Affiliation(s)
- Bani Kumar Pathak
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sandip Dey
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sukanya Mozumder
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jayati Sengupta
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
38
|
Essential contributions of food hydrocolloids and phospholipid liposomes to the formation of carriers for controlled delivery of biologically active substances via the gastrointestinal tract. Food Hydrocoll 2021. [DOI: 10.1016/j.foodhyd.2021.106890] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
Lorentzon E, Horvath I, Kumar R, Rodrigues JI, Tamás MJ, Wittung-Stafshede P. Effects of the Toxic Metals Arsenite and Cadmium on α-Synuclein Aggregation In Vitro and in Cells. Int J Mol Sci 2021; 22:ijms222111455. [PMID: 34768886 PMCID: PMC8584132 DOI: 10.3390/ijms222111455] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 11/21/2022] Open
Abstract
Exposure to heavy metals, including arsenic and cadmium, is associated with neurodegenerative disorders such as Parkinson’s disease. However, the mechanistic details of how these metals contribute to pathogenesis are not well understood. To search for underlying mechanisms involving α-synuclein, the protein that forms amyloids in Parkinson’s disease, we here assessed the effects of arsenic and cadmium on α-synuclein amyloid formation in vitro and in Saccharomyces cerevisiae (budding yeast) cells. Atomic force microscopy experiments with acetylated human α-synuclein demonstrated that amyloid fibers formed in the presence of the metals have a different fiber pitch compared to those formed without metals. Both metal ions become incorporated into the amyloid fibers, and cadmium also accelerated the nucleation step in the amyloid formation process, likely via binding to intermediate species. Fluorescence microscopy analyses of yeast cells expressing fluorescently tagged α-synuclein demonstrated that arsenic and cadmium affected the distribution of α-synuclein aggregates within the cells, reduced aggregate clearance, and aggravated α-synuclein toxicity. Taken together, our in vitro data demonstrate that interactions between these two metals and α-synuclein modulate the resulting amyloid fiber structures, which, in turn, might relate to the observed effects in the yeast cells. Whilst our study advances our understanding of how these metals affect α-synuclein biophysics, further in vitro characterization as well as human cell studies are desired to fully appreciate their role in the progression of Parkinson’s disease.
Collapse
Affiliation(s)
- Emma Lorentzon
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (E.L.); (J.I.R.)
| | - Istvan Horvath
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden; (I.H.); (R.K.)
| | - Ranjeet Kumar
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden; (I.H.); (R.K.)
| | - Joana Isabel Rodrigues
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (E.L.); (J.I.R.)
| | - Markus J. Tamás
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (E.L.); (J.I.R.)
- Correspondence: (M.J.T.); (P.W.-S.)
| | - Pernilla Wittung-Stafshede
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden; (I.H.); (R.K.)
- Correspondence: (M.J.T.); (P.W.-S.)
| |
Collapse
|
40
|
Dehghani M, Jalal R, Rashidi MR. Kinetic and thermodynamic insights into the interaction of Aβ1-42 with astaxanthin and aggregation behavior of Aβ1-42: Surface plasmon resonance, microscopic, and molecular docking studies. Biophys Chem 2021; 275:106612. [PMID: 33984664 DOI: 10.1016/j.bpc.2021.106612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/18/2022]
Abstract
Amyloid-β 1-42 (Aβ1-42) aggregation is considered as an important process in the pathology of Alzheimer's disease (AD). Astaxanthin (ATX), a xanthophyll carotenoid, has a broad range of biological activities such as neuroprotective one. The present study aimed to elucidate the interaction of ATX with Aβ1-42, as well as its effect on Aβ1-42 aggregates under different conditions. Based on the surface plasmon resonance (SPR) results, ATX possessed a high affinity towards Aβ1-42 and the binding process was spontaneous, endothermic, and entropy-driven. Additionally, the binding affinity of ATX to Aβ1-42 was glucose and insulin concentration-dependent. Hydrophobic interactions may play an important role in the interaction between ATX and Aβ1-42. The results of SPR, thioflavin T (ThT), and transmission electron microscopy (TEM) analyses represented the dependency of the anti-amyloid activity of ATX on glucose, insulin, and ATX concentrations. Further, molecular docking results indicated the presence of some same binding sites on Aβ1-42 for ATX and glucose, as well as ATX and insulin, which suggests the possible competition between the molecules for Aβ1-42 binding. Furthermore, the MTT results confirmed that ATX effect on the viability of Aβ1-42-treated PC12 cells was dependent on glucose, insulin, and ATX concentrations. In general, the results provided further insights into the interaction between Aβ1-42 and ATX, as well as the effect of ATX on Aβ1-42 aggregates under various conditions.
Collapse
Affiliation(s)
- Moharram Dehghani
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Razieh Jalal
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mohammad-Reza Rashidi
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Tan Q, Liu H, Duan M, Huo S. Interplay between human islet amyloid polypeptide aggregates and micro-heterogeneous membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183691. [PMID: 34224702 DOI: 10.1016/j.bbamem.2021.183691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Human islet amyloid polypeptides (hIAPP) aggregate into amyloid deposits in the pancreatic islets of Langerhans, contributing to the loss of β-cells of patients with type 2 diabetes. Despite extensive studies of membrane disruption associated with hIAPP aggregates, the molecular details regarding the complex interplay between hIAPP aggregates and raft-containing membranes are still very limited. Using all-atom molecular dynamics simulations, we investigate the impact of hIAPP aggregate insertion on lipid segregation. We have found that the domain separation of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) is enhanced upon hIAPP membrane permeabilization in the absence of cholesterol, while within our simulation timescale, we cannot provide definitive evidence regarding the impact of hIAPP insertion on domain segregation in the ternary mixture (DOPC/DPPC/cholesterol). When the lipid domains are perturbed, their restoration occurs rapidly and spontaneously in the presence of hIAPP aggregates. hIAPP insertion affects membrane thickness in its immediate surroundings. On average, hIAPP causes the fluidity of lipids to increase and even cholesterol shows enhanced diffusivity. The acyl chain packing of the lipids near hIAPP is disrupted as compared to that further away from it. Cholesterol not only modulates membrane mobility and ordering but also hIAPP aggregates' structure and relative orientation to the membrane. Our investigations on the interaction between hIAPP aggregates and raft-containing membranes could lead to a better understanding of the mechanisms of amyloid cytotoxicity.
Collapse
Affiliation(s)
- Qingzhe Tan
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, 950 Main Street, Worcester, MA 01610, USA
| | - Hanzhong Liu
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, 950 Main Street, Worcester, MA 01610, USA
| | - Mojie Duan
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, 950 Main Street, Worcester, MA 01610, USA; Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Shuanghong Huo
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, 950 Main Street, Worcester, MA 01610, USA.
| |
Collapse
|
42
|
C subunit of the ATP synthase is an amyloidogenic calcium dependent channel-forming peptide with possible implications in mitochondrial permeability transition. Sci Rep 2021; 11:8744. [PMID: 33888826 PMCID: PMC8062469 DOI: 10.1038/s41598-021-88157-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/06/2021] [Indexed: 01/18/2023] Open
Abstract
The c subunit is an inner mitochondrial membrane (IMM) protein encoded by three nuclear genes. Best known as an integral part of the F0 complex of the ATP synthase, the c subunit is also present in other cytoplasmic compartments in ceroid lipofuscinoses. Under physiological conditions, this 75 residue-long peptide folds into an α-helical hairpin and forms oligomers spanning the lipid bilayer. In addition to its physiological role, the c subunit has been proposed as a key participant in stress-induced IMM permeabilization by the mechanism of calcium-induced permeability transition. However, the molecular mechanism of the c subunit participation in IMM permeabilization is not completely understood. Here we used fluorescence spectroscopy, atomic force microscopy and black lipid membrane methods to gain insights into the structural and functional properties of unmodified c subunit protein that might make it relevant to mitochondrial toxicity. We discovered that c subunit is an amyloidogenic peptide that can spontaneously fold into β-sheets and self-assemble into fibrils and oligomers in a Ca2+-dependent manner. C subunit oligomers exhibited ion channel activity in lipid membranes. We propose that the toxic effects of c subunit might be linked to its amyloidogenic properties and are driven by mechanisms similar to those of neurodegenerative polypeptides such as Aβ and α-synuclein.
Collapse
|
43
|
Abstract
Protein aggregation and amyloid formation are pathogenic events underlying the development of an increasingly large number of human diseases named “proteinopathies”. Abnormal accumulation in affected tissues of amyloid β (Aβ) peptide, islet amyloid polypeptide (IAPP), and the prion protein, to mention a few, are involved in the occurrence of Alzheimer’s (AD), type 2 diabetes mellitus (T2DM) and prion diseases, respectively. Many reports suggest that the toxic properties of amyloid aggregates are correlated with their ability to damage cell membranes. However, the molecular mechanisms causing toxic amyloid/membrane interactions are still far to be completely elucidated. This review aims at describing the mutual relationships linking abnormal protein conformational transition and self-assembly into amyloid aggregates with membrane damage. A cross-correlated analysis of all these closely intertwined factors is thought to provide valuable insights for a comprehensive molecular description of amyloid diseases and, in turn, the design of effective therapies.
Collapse
|
44
|
Effect of phospholipids on the physicochemical properties of myofibrillar proteins solution mediated by NaCl concentration. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.110895] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
FoxA2 and RNA Pol II mediate human islet amyloid polypeptide turnover in ER-stressed pancreatic β-cells. Biochem J 2021; 478:1261-1282. [PMID: 33650632 DOI: 10.1042/bcj20200984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/25/2022]
Abstract
Here, we investigated transcriptional and trafficking mechanisms of human islet amyloid polypeptide (hIAPP) in normal and stressed β-cells. In high glucose-challenged human islets and rat insulinoma cells overexpressing hIAPP, cell fractionation studies revealed increased accumulation of hIAPP. Unexpectedly, a significant fraction (up to 22%) of hIAPP was found in the nuclear soluble and chromatin-enriched fractions of cultured human islet and rat insulinoma cells. The nucleolar accumulation of monomeric forms of hIAPP did not have any adverse effect on the proliferation of β-cells nor did it affect nucleolar organization or function. However, intact nucleolar organization and function were essential for hIAPP expression under normal and ER-stress conditions as RNA polymerase II inhibitor, α-amanitin, reduced hIAPP protein expression evoked by high glucose and thapsigargin. Promoter activity studies revealed the essential role of transcription factor FoxA2 in hIAPP promoter activation in ER-stressed β-cells. Transcriptome and secretory studies demonstrate that the biosynthetic and secretory capacity of islet β-cells was preserved during ER stress. Thus, the main reason for increased intracellular hIAPP accumulation is its enhanced biosynthesis under these adverse conditions.
Collapse
|
46
|
Gonzalez-Garcia M, Fusco G, De Simone A. Membrane Interactions and Toxicity by Misfolded Protein Oligomers. Front Cell Dev Biol 2021; 9:642623. [PMID: 33791300 PMCID: PMC8006268 DOI: 10.3389/fcell.2021.642623] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/08/2021] [Indexed: 01/13/2023] Open
Abstract
The conversion of otherwise soluble proteins into insoluble amyloid aggregates is associated with a range of neurodegenerative disorders, including Alzheimer’s and Parkinson’s diseases, as well as non-neuropathic conditions such as type II diabetes and systemic amyloidoses. It is increasingly evident that the most pernicious species among those forming during protein aggregation are small prefibrillar oligomers. In this review, we describe the recent progress in the characterization of the cellular and molecular interactions by toxic misfolded protein oligomers. A fundamental interaction by these aggregates involves biological membranes, resulting in two major model mechanisms at the onset of the cellular toxicity. These include the membrane disruption model, resulting in calcium imbalance, mitochondrial dysfunction and intracellular reactive oxygen species, and the direct interaction with membrane proteins, leading to the alteration of their native function. A key challenge remains in the characterization of transient interactions involving heterogeneous protein aggregates. Solving this task is crucial in the quest of identifying suitable therapeutic approaches to suppress the cellular toxicity in protein misfolding diseases.
Collapse
Affiliation(s)
- Mario Gonzalez-Garcia
- Department of Life Sciences, Imperial College London, South Kensington, United Kingdom
| | - Giuliana Fusco
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Alfonso De Simone
- Department of Life Sciences, Imperial College London, South Kensington, United Kingdom.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
47
|
García-Viñuales S, Sciacca MFM, Lanza V, Santoro AM, Grasso G, Tundo GR, Sbardella D, Coletta M, Grasso G, La Rosa C, Milardi D. The interplay between lipid and Aβ amyloid homeostasis in Alzheimer's Disease: risk factors and therapeutic opportunities. Chem Phys Lipids 2021; 236:105072. [PMID: 33675779 DOI: 10.1016/j.chemphyslip.2021.105072] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/15/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022]
Abstract
Alzheimer's Diseases (AD) is characterized by the accumulation of amyloid deposits of Aβ peptide in the brain. Besides genetic background, the presence of other diseases and an unhealthy lifestyle are known risk factors for AD development. Albeit accumulating clinical evidence suggests that an impaired lipid metabolism is related to Aβ deposition, mechanistic insights on the link between amyloid fibril formation/clearance and aberrant lipid interactions are still unavailable. Recently, many studies have described the key role played by membrane bound Aβ assemblies in neurotoxicity. Moreover, it has been suggested that a derangement of the ubiquitin proteasome pathway and autophagy is significantly correlated with toxic Aβ aggregation and dysregulation of lipid levels. Thus, studies focusing on the role played by lipids in Aβ aggregation and proteostasis could represent a promising area of investigation for the design of valuable treatments. In this review we examine current knowledge concerning the effects of lipids in Aβ aggregation and degradation processes, focusing on the therapeutic opportunities that a comprehensive understanding of all biophysical, biochemical, and biological processes involved may disclose.
Collapse
Affiliation(s)
| | - Michele F M Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Valeria Lanza
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Anna Maria Santoro
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Giulia Grasso
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Grazia R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Massimiliano Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Grasso
- Department of Chemistry, University of Catania, Catania, Italy
| | - Carmelo La Rosa
- Department of Chemistry, University of Catania, Catania, Italy
| | - Danilo Milardi
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy.
| |
Collapse
|
48
|
Effect of packing density of lipid vesicles on the Aβ42 fibril polymorphism. Chem Phys Lipids 2021; 236:105073. [PMID: 33675780 DOI: 10.1016/j.chemphyslip.2021.105073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 11/22/2022]
Abstract
The aggregation of amyloid-β 1-42 (Aβ42) on lipid membranes is closely related to the pathology of Alzheimer's disease (AD). Herein, we demonstrated the effect of the packing density of lipid vesicles on the Aβ42 fibrillation kinetics and fibril morphology. We used three distinct phosphatidylcholine (PC) lipids, containing different numbers of cis-double bonds in acyl chains, and therefore, a different packing density in the lipid vesicles. Our results showed that the fibrillation of Aβ42 was greatly enhanced and the formed fibrils became shorter as the number of double bonds in lipids increased. Due to the low-density characteristics of dioleoyl phosphatidylcholine (DOPC), Aβ42 monomers were able to interact with the hydrophobic acyl chain of lipids exposed to the aqueous phase, thereby inducing rapid fibrillation and short fibril morphologies. Furthermore, the effects of the anionic lipids dioleoyl phosphatidylserine (DOPS) and dioleoyl phosphatidylglycerol (DOPG), and mixed vesicles of DOPC/DOPS and DOPC/DOPG on Aβ42 fibrillations were investigated. The tight binding of Aβ42 to the lipid head groups via electrostatic interactions was able to suppress the modulation of Aβ42 fibrillations compared to accelerated fibrillations on loosely packed membranes. Our proposed mechanism regarding the influence of lipid packing density on Aβ42 fibrillations provides an advanced understanding of lipid-associated amyloid fibrillations.
Collapse
|
49
|
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: 117] [Impact Index Per Article: 29.3] [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
|
50
|
Ugbaja SC, Sanusi ZK, Appiah-Kubi P, Lawal MM, Kumalo HM. Computational modelling of potent β-secretase (BACE1) inhibitors towards Alzheimer's disease treatment. Biophys Chem 2020; 270:106536. [PMID: 33387910 DOI: 10.1016/j.bpc.2020.106536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/28/2022]
Abstract
Researchers have identified the β-amyloid precursor protein cleaving enzyme 1 (BACE1) in the multifactorial pathway of Alzheimer's disease (AD) as a drug target. The design and development of molecules to inhibit BACE1 as a potential cure for AD thus remained significant. Herein, we simulated two potent BACE1 inhibitors (AM-6494 and CNP-520) to understand their binding affinity at the atomistic level. AM-6494 is a newly reported potent BACE1 inhibitor with an IC50 value of 0.4 nM in vivo and now picked for preclinical considerations. Umibecestat (CNP-520), which was discontinued at human trials lately, was considered to enable a reasonable evaluation of our results. Using density functional theory (DFT) and Our Own N-layered Integrated molecular Orbital and Molecular Mechanics (ONIOM), we achieved the aim of this investigation. These computational approaches enabled the prediction of the electronic properties of AM-6494 and CNP-520 plus their binding energies when complexed with BACE1. For AM-6494 and CNP-520 interaction with protonated BACE1, the ONIOM calculation gave binding free energy of -62.849 and -33.463 kcal/mol, respectively. In the unprotonated model, we observed binding free energy of -59.758 kcal/mol in AM-6494. Taken together thermochemistry of the process and molecular interaction plot, AM-6494 is more favourable than CNP-520 towards the inhibition of BACE1. The protonated model gave slightly better binding energy than the unprotonated form. However, both models could sufficiently describe ligand binding to BACE1 at the atomistic level. Understanding the detailed molecular interaction of these inhibitors could serve as a basis for pharmacophore exploration towards improved inhibitor design.
Collapse
Affiliation(s)
- Samuel C Ugbaja
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Zainab K Sanusi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Patrick Appiah-Kubi
- Molecular Bio-computational and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Monsurat M Lawal
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa.
| | - Hezekiel M Kumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa.
| |
Collapse
|