1
|
Devi S, Garg DK, Bhat R. Green tea polyphenol EGCG acts differentially on end-stage amyloid polymorphs of α-synuclein formed in different polyol osmolytes. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141073. [PMID: 40189174 DOI: 10.1016/j.bbapap.2025.141073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/10/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Synucleinopathies are heterogenous group of disorders characterized by α-synuclein amyloid aggregates in the nervous system. Different synucleinopathy clinical subtypes are encoded by structurally diverse α-synuclein amyloid polymorphs referred to as 'strains'. The underlying structural differences between polymorphs can potentially hamper the drug design against synucleinopathies. Polyphenolic compounds like EGCG have shown promise in inhibiting and remodeling of α-synuclein amyloid aggregates, but their effects on different polymorphs are not well-studied. The cellular environment is one factor contributing to the heterogeneity in the amyloid landscape. Herein, we generated diverse polymorphs of α-synuclein by fine-tuning its aggregation using different polyol osmolytes, varying in their physicochemical properties. These osmolytes act as globular protein stabilizers and conformational modulators of intrinsically disordered proteins. While the buffer control α-synuclein aggregates were evenly dispersed, the polyol-induced aggregate solutions contained a heterogeneous mixture of co-existing polymorphs, as evidenced by AFM and TEM measurements. The polyol-induced aggregated solutions consisted of a mixture of both fibrillar and nonfibrillar cross-β-rich species. Using various spectroscopic tools, we observed differences in the structures of osmolyte-induced polymorphic aggregates. We incubated these aggregates with EGCG and observed its disparate action over polymorphs wherein the treated species were either disintegrated or structurally altered. Contrary to previous reports, all EGCG-treated polymorphs were β-sheet-rich and seeding-competent. Our findings are relevant in assessing the efficacy of polyphenolic compounds on diverse aggregate strains encoding different proteinopathy variants. The formation of β-sheet-rich species in our study also engenders a more critical examination of EGCG's mode of action on diverse classes of amyloids.
Collapse
Affiliation(s)
- Santosh Devi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Dushyant K Garg
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India; Department of Biomedicine, University of Bergen 5009, Norway
| | - Rajiv Bhat
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
2
|
Farrokhzad R, Seyedalipour B, Baziyar P, Hosseinkhani S. Insight Into Factors Influencing the Aggregation Process in Wild-Type and P66R Mutant SOD1: Computational and Spectroscopic Approaches. Proteins 2025; 93:885-907. [PMID: 39643934 DOI: 10.1002/prot.26765] [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: 08/03/2024] [Revised: 10/02/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
Disturbances in metal ion homeostasis associated with amyotrophic lateral sclerosis (ALS) have been described for several years, but the exact mechanism of involvement is not well understood. To elucidate the role of metalation in superoxide dismutase (SOD1) misfolding and aggregation, we comprehensively characterized the structural features (apo/holo forms) of WT-SOD1 and P66R mutant in loop IV. Using computational and experimental methodologies, we assessed the physicochemical properties of these variants and their correlation with protein aggregation at the molecular level. Modifications in apo-SOD1 compared to holo-SOD1 were more pronounced in flexibility, stability, hydrophobicity, and intramolecular interactions, as indicated by molecular dynamics simulations. The enzymatic activities of holo/apo-WT SOD1 were 1.30 and 1.88-fold of the holo/apo P66R mutant, respectively. Under amyloid-inducing conditions, decreased ANS fluorescence intensity in the apo-form relative to the holo-form suggested pre-fibrillar species and amyloid aggregate growth due to occluded hydrophobic pockets. FTIR spectroscopy revealed that apo-WT-SOD1 and apo-P66R exhibited a mixture of parallel and intermolecular β-sheet structures, indicative of aggregation propensity. Aggregate species were identified using TEM, Congo red staining, and ThT/ANS fluorescence spectroscopy. Thermodynamic analyses with GdnHCl demonstrated that metal deficit, mutation, and intramolecular disulfide bond reduction are essential for initiating SOD1 misfolding and aggregation. These disruptions destabilize the dimer-monomer equilibrium, promoting dimer dissociation into monomers and decreasing the thermodynamic stability of SOD1 variants, thus facilitating amyloid/amorphous aggregate formation. Our findings offer novel insights into protein aggregation mechanisms in disease pathology and highlight potential therapeutic strategies against toxic protein aggregation, including SOD1.
Collapse
Affiliation(s)
- Roghayeh Farrokhzad
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Bagher Seyedalipour
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Payam Baziyar
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Bencs F, Románszki L, Farkas V, Perczel A. Structural Insights Into Amyloid Polymorphism: The Impact of Glutamine to Norleucine Substitutions in GNNQQNY Aggregation. Chemistry 2025:e202404255. [PMID: 40152416 DOI: 10.1002/chem.202404255] [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: 11/18/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 03/29/2025]
Abstract
Polypeptides can self-assemble into highly organized amyloid structures through complex and poorly understood mechanisms. To better understand the key parameters governing amyloidogenesis, we investigated the aggregation of the Sup35 prion-derived GNNQQNY sequence alongside two rationally designed mutants, glutamine to norleucine in the 4th or 5th position, where selective removal of hydrogen bonding capacity reduces amyloid structural stability. Our findings reveal that β-sheet arrays form rapidly as an initial step, followed by π-π aromatic interactions between Tyr residues, which drive hierarchical self-assembly into 3D fibrillar structures via hydrophobic zippers and partial water exclusion. As the oligomers grow, they also acquire twist and chirality at the protofilament level, with Tyr ladders serving as key interaction surfaces that dictate the final amyloid architecture. These ladders guide protofibrils to assemble into either oppositely twisted chiral fibers or achiral nanocrystals, contributing to amyloid polymorphism. The emergence of distinct polymorphs is influenced by multiple factors, including fibril twisting, side-chain interactions, solvent exclusion, and local microenvironmental conditions. Our study provides crucial insights into the hierarchical nature of amyloid self-assembly and highlights the structural adaptability of amyloid fibrils, which is essential for designing functional amyloids and understanding the pathogenicity of disease-associated aggregates.
Collapse
Affiliation(s)
- Fruzsina Bencs
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, Budapest, Hungary
- ELTE Hevesy György PhD School of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, Budapest, Hungary
| | - Loránd Románszki
- HUN-REN Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, Magyar tudósok körútja 2, Budapest, Hungary
| | - Viktor Farkas
- HUN-REN - ELTE Protein Modeling Research Group, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, Budapest, Hungary
| | - András Perczel
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, Budapest, Hungary
- HUN-REN - ELTE Protein Modeling Research Group, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, Budapest, Hungary
| |
Collapse
|
4
|
Wang X, Mondal M, Jankoski PE, Kemp LK, Clemons TD, Rangachari V, Morgan SE. De Novo Amyloid Peptide-Polymer Blends with Enhanced Mechanical and Biological Properties. ACS APPLIED POLYMER MATERIALS 2025; 7:3739-3751. [PMID: 40177395 PMCID: PMC11959523 DOI: 10.1021/acsapm.4c04020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025]
Abstract
Amyloid peptides are structurally diverse materials that exhibit different properties depending on their self-assembly. While they are often associated with neurodegenerative diseases, functional amyloids play important roles in nature and exhibit properties with high relevance for biomedical applications, including remarkable strength, mechanical stability, antimicrobial and antioxidant properties, low cytotoxicity, and adhesion to biotic and abiotic surfaces. Challenges in developing amyloid biomaterials include the complexity of peptide chemistry and the practical techniques required for processing amyloids into bulk materials. In this work, two de novo decapeptides with fibrillar and globular morphologies were synthesized, blended with poly(ethylene oxide), and fabricated into composite mats via electrospinning. Notable enhancements in the mechanical properties of the composite mats were observed, attributed to the uniform distribution of the peptide assemblies within the PEO matrix and interactions between the materials. Morphological differences, such as the production of thinner nanofibers, are attributed to the increased conductivity from the zwitterionic nature of the decapeptides. Blend rheology and postprocessing analysis revealed how processing might affect the amyloid aggregation and secondary structure of the peptides. Both decapeptides demonstrated low cytotoxicity and strong antioxidant activity, indicating their potential for safe and effective use as biomaterials. This research lays the foundation for designing amyloid peptides for specific applications by defining the structure-property-processing relationships of the de novo peptide-polymer blends.
Collapse
Affiliation(s)
- Xianjun Wang
- School of
Polymer Science and Engineering, University
of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Malay Mondal
- Department
of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Penelope E. Jankoski
- School of
Polymer Science and Engineering, University
of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Lisa K. Kemp
- School of
Polymer Science and Engineering, University
of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Tristan D. Clemons
- School of
Polymer Science and Engineering, University
of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
- Center
for
Molecular and Cellular Biosciences, University
of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Vijayaraghavan Rangachari
- Department
of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
- Center
for
Molecular and Cellular Biosciences, University
of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Sarah E. Morgan
- School of
Polymer Science and Engineering, University
of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| |
Collapse
|
5
|
Kardos J, Nyiri MP, Moussong É, Wien F, Molnár T, Murvai N, Tóth V, Vadászi H, Kun J, Jamme F, Micsonai A. Guide to the structural characterization of protein aggregates and amyloid fibrils by CD spectroscopy. Protein Sci 2025; 34:e70066. [PMID: 39968955 PMCID: PMC11836901 DOI: 10.1002/pro.70066] [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: 08/31/2024] [Revised: 12/21/2024] [Accepted: 02/01/2025] [Indexed: 02/20/2025]
Abstract
Protein aggregation and amyloid formation are linked to numerous degenerative diseases, such as Alzheimer's or Parkinson's disease. Additionally, protein aggregation plays a crucial role in various biological processes, such as storage of molecules or cell signaling. Protein molecules can form a wide range of aggregates, from oligomers of different sizes to non-specific aggregates and highly ordered cross-β structured amyloid fibrils with diverse morphologies. Circular dichroism (CD) spectroscopy is a widely used technique to study protein structures providing detailed information at the secondary structure level, and is ideal to distinguish and characterize protein aggregates. Despite its potential, CD spectroscopy is often perceived as having limited application on protein aggregates due to challenges, such as sample inhomogeneity, precipitation, light scattering and other factors that complicate accurate analysis. In this study, we present a detailed protocol for examining the structure of protein aggregates and amyloid fibrils using CD spectroscopy. We outline the optimal experimental conditions for sample preparation and demonstrate how to identify and mitigate various interfering effects, using specific examples of disease-related amyloidogenic proteins. We also discuss the instrumental parameters, baseline subtraction, normalization, and quality control of CD spectra. Furthermore, we evaluate the performance of different secondary structure estimating algorithms on amyloid fibril CD spectra highlighting the superiority of BeStSel and CDNN. Our findings could enhance the structural analysis of protein aggregates, contributing to a better understanding of associated diseases and the development of new therapeutic strategies.
Collapse
Affiliation(s)
- József Kardos
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Márton Péter Nyiri
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Éva Moussong
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
- ELTE—Functional Nucleic Acid Motifs Research Group, Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Frank Wien
- DISCO BeamlineSynchrotron SOLEILGif‐sur‐YvetteFrance
| | - Tamás Molnár
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Nikoletta Murvai
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
- ELTE—Functional Nucleic Acid Motifs Research Group, Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Vilmos Tóth
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Henrietta Vadászi
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Judit Kun
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| | | | - András Micsonai
- Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
- ELTE—Functional Nucleic Acid Motifs Research Group, Department of Biochemistry, Institute of BiologyELTE Eötvös Loránd UniversityBudapestHungary
| |
Collapse
|
6
|
Lakela AL, Berntsson E, Vosough F, Jarvet J, Paul S, Barth A, Gräslund A, Roos PM, Wärmländer SKTS. Molecular interactions, structural effects, and binding affinities between silver ions (Ag +) and amyloid beta (Aβ) peptides. Sci Rep 2025; 15:5439. [PMID: 39948350 PMCID: PMC11825922 DOI: 10.1038/s41598-024-59826-6] [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: 12/05/2023] [Accepted: 04/16/2024] [Indexed: 02/16/2025] Open
Abstract
Because silver is toxic to microbes, but not considered toxic to humans, the metal has been used as an antimicrobial agent since ancient times. Today, silver nanoparticles and colloidal silver are used for antibacterial purposes, and silver-peptide and similar complexes are being developed as therapeutic agents. Yet, the health effects of silver exposure are not fully understood, nor are the molecular details of silver-protein interactions. In Alzheimer's disease, the most common form of dementia worldwide, amyloid-β (Aβ) peptides aggregate to form soluble oligomers that are neurotoxic. Here, we report that monovalent silver ions (Ag+) bind wildtype Aβ40 peptides with a binding affinity of 25 ± 12 µM in MES buffer at 20 °C. Similar binding affinities are observed for wt Aβ40 peptides bound to SDS micelles, for an Aβ40(H6A) mutant, and for a truncated Aβ(4-40) variant containing an ATCUN (Amino Terminal Cu and Ni) motif. Weaker Ag+ binding is observed for the wt Aβ40 peptide at acidic pH, and for an Aβ40 mutant without histidines. These results are compatible with Ag+ ions binding to the N-terminal segment of Aβ peptides with linear bis-his coordination. Because the Ag+ ions do not induce any changes in the size or structure of Aβ42 oligomers, we suggest that Ag+ ions have a minor influence on Aβ toxicity.
Collapse
Affiliation(s)
- Amanda L Lakela
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden
| | - Elina Berntsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden
- CellPept Sweden AB, Kvarngatan 10B, 11847, Stockholm, Sweden
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 19086, Tallinn, Estonia
| | - Faraz Vosough
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden
- CellPept Sweden AB, Kvarngatan 10B, 11847, Stockholm, Sweden
- The National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Suman Paul
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden
| | - Andreas Barth
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden.
- CellPept Sweden AB, Kvarngatan 10B, 11847, Stockholm, Sweden.
| | - Per M Roos
- Institute of Environmental Medicine, Karolinska Institutet, 17177, Stockholm, Sweden
- University Healthcare Unit of Capio St. Göran Hospital, 11281, Stockholm, Sweden
| | - Sebastian K T S Wärmländer
- CellPept Sweden AB, Kvarngatan 10B, 11847, Stockholm, Sweden.
- Chemistry Section, Arrhenius Laboratories, Stockholm University, 10691, Stockholm, Sweden.
| |
Collapse
|
7
|
Bilog M, Cersosimo J, Vigil I, Desamero RZB, Profit AA. Effect of a SARS-CoV-2 Protein Fragment on the Amyloidogenic Propensity of Human Islet Amyloid Polypeptide. ACS Chem Neurosci 2024; 15:4431-4440. [PMID: 39582236 PMCID: PMC11660541 DOI: 10.1021/acschemneuro.4c00473] [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: 07/25/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024] Open
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the onset of COVID-19 have been linked to an increased risk of developing type 2 diabetes. While a variety of mechanisms may ultimately be responsible for the onset of type 2 diabetes under these circumstances, one mechanism that has been postulated involves the increased aggregation of human islet amyloid polypeptide (hIAPP) through direct interaction with SARS-CoV-2 viral proteins. Previous computational studies investigating this possibility revealed that a nine-residue peptide fragment known as SK9 (SFYVYSRVK) from the SARS-CoV-2 envelope protein can stabilize the native conformation of hIAPP1-37 by interacting with the N-terminal region of amylin. One of the areas particularly stabilized through this interaction encompasses residues 15-28 of amylin. Given these findings, we investigated whether SK9 could interact with short amyloidogenic sequences derived from this region of amylin. Here, we employ docking studies, molecular dynamics simulations, and biophysical techniques to provide theoretical as well as direct experimental evidence that SK9 can interact with hIAPP12-18 and hIAPP20-29 peptides. Furthermore, we demonstrate that SK9 not only can interact with these sequences but also serves to prevent the self-assembly of these amyloidogenic peptides. In striking contrast, we also show that SK9 has little effect on the amyloidogenic propensity of full-length amylin. These findings are contrary to previous published simulations involving SK9 and hIAPP1-37. Such observations may assist in clarifying potential mechanisms of the SARS-CoV-2 interaction with hIAPP and its relevance to the onset of type 2 diabetes in the setting of COVID-19.
Collapse
Affiliation(s)
- Marvin Bilog
- PhD
Programs in Chemistry and Biochemistry, the Graduate Center of the
City University of New York, New
York, New York 10016, United States
- Department
of Chemistry, York College of the City University
of New York, Jamaica, New York 11451, United States
| | - Jennifer Cersosimo
- PhD
Programs in Chemistry and Biochemistry, the Graduate Center of the
City University of New York, New
York, New York 10016, United States
- Department
of Chemistry, York College of the City University
of New York, Jamaica, New York 11451, United States
| | - Iliana Vigil
- Department
of Chemistry, York College of the City University
of New York, Jamaica, New York 11451, United States
| | - Ruel Z. B. Desamero
- PhD
Programs in Chemistry and Biochemistry, the Graduate Center of the
City University of New York, New
York, New York 10016, United States
- Department
of Chemistry, York College of the City University
of New York, Jamaica, New York 11451, United States
| | - Adam A. Profit
- PhD
Programs in Chemistry and Biochemistry, the Graduate Center of the
City University of New York, New
York, New York 10016, United States
- Department
of Chemistry, York College of the City University
of New York, Jamaica, New York 11451, United States
| |
Collapse
|
8
|
Bhattacharya I, Saha R, Pyne S, Bera A, Mitra RK. Excipient Induced Unusual Phase Separation in Bovine Serum Albumin Solution: An Explicit Role Played by Ion-Hydration. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:25822-25833. [PMID: 39575889 DOI: 10.1021/acs.langmuir.4c02802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
We report an instantaneous room-temperature phase separation of 1 mM bovine serum albumin solution in the presence of (20% acetic acid+0.2 M NaCl), a routinely used food preservative; an opaque liquid-like phase (L) coexists in equilibrium with a granular gel like phase (G). Interestingly, neither 20% acetic acid nor 0.2 M NaCl individually induces such a phase separation. Field emission scanning electron microscopy (FESEM) and high-resolution transmission electron microscopy (HRTEM) imaging show aggregated proteins to be dispersed in the upper phase, while the lower phase is composed of cross-linked fibrils (hydrogels). Mid-IR FTIR, Raman scattering, and circular dichroism (CD) experiments reveal a significant increase in the β-sheet content in BSA, which confirms the formation of amyloids in the presence of the excipient. Both L and G phases undergo distinct visual and microscopic changes upon incubation at 25 and 80 °C. It is evident that the added salt plays a pivotal role in bringing about this otherwise unique phase behavior. We divulge the explicit role of the ion associated hydration using THz-FTIR measurements in the 1.5-16.7 THz (50-550 cm-1) frequency window. Systematic alteration in the ion-induced THz-active mode of water envisions the key role of ions in shaping the various phases. Our study depicts an intriguing observation of severe amyloidosis of BSA upon the addition of a food preservative, even at room temperature, which is expected to add new insight into amyloid research. Considering the increasing number of individuals suffering from several neurodegenerative disorders (Alzheimer's, Parkinson's, type-2 diabetes, obesity, cancer, etc.), this study leads a caution toward critically revisiting the usage of known food preservatives.
Collapse
Affiliation(s)
- Indrani Bhattacharya
- Department of Chemical and Biological Sciences S.N. Bose National Centre for Basic Sciences Block-JD, Sector-III, Salt Lake Kolkata-700106, India
| | - Ria Saha
- Department of Chemical and Biological Sciences S.N. Bose National Centre for Basic Sciences Block-JD, Sector-III, Salt Lake Kolkata-700106, India
| | - Sumana Pyne
- Department of Chemical and Biological Sciences S.N. Bose National Centre for Basic Sciences Block-JD, Sector-III, Salt Lake Kolkata-700106, India
| | - Asesh Bera
- Department of Chemical and Biological Sciences S.N. Bose National Centre for Basic Sciences Block-JD, Sector-III, Salt Lake Kolkata-700106, India
| | - Rajib Kumar Mitra
- Department of Chemical and Biological Sciences S.N. Bose National Centre for Basic Sciences Block-JD, Sector-III, Salt Lake Kolkata-700106, India
| |
Collapse
|
9
|
Huang SH, Fang ST, Yang CH, Liou JW, Chen YC. Modulating Amyloid-β Toxicity: In Vitro Analysis of Aβ42(G37V) Variant Impact on Aβ42 Aggregation and Cytotoxicity. Int J Mol Sci 2024; 25:13219. [PMID: 39684928 DOI: 10.3390/ijms252313219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is primarily driven by the formation of toxic amyloid-β (Aβ) aggregates, with Aβ42 being a pivotal contributor to disease pathology. This study investigates a novel agent to mitigate Aβ42-induced toxicity by co-assembling Aβ42 with its G37V variant (Aβ42(G37V)), where Gly at position 37 is substituted with valine. Using a combination of Thioflavin-T (Th-T) fluorescence assays, Western blot analysis, atomic force microscopy (AFM)/transmission electron microscopy (TEM), and biochemical assays, we demonstrated that adding Aβ42(G37V) significantly accelerates Aβ42 aggregation rate and mass while altering the morphology of the resulting aggregates. Consequently, adding Aβ42(G37V) reduces the Aβ42 aggregates-induced cytotoxicity, as evidenced by improved cell viability assays. The possible mechanism for this effect is that adding Aβ42(G37V) reduces the production of reactive oxygen species (ROS) and lipid peroxidation, typically elevated in response to Aβ42, indicating its protective effects against oxidative stress. These findings suggest that Aβ42(G37V) could be a promising candidate for modulating Aβ42 aggregation dynamics and reducing its neurotoxic effects, providing a new avenue for potential therapeutic interventions in AD.
Collapse
Affiliation(s)
- Shu-Hsiang Huang
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Shang-Ting Fang
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Chin-Hao Yang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City 970, Taiwan
| | - Je-Wen Liou
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City 970, Taiwan
| | - Yi-Cheng Chen
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| |
Collapse
|
10
|
Siri M, Vázquez-Dávila M, Sotelo Guzman C, Bidan CM. Nutrient availability influences E. coli biofilm properties and the structure of purified curli amyloid fibers. NPJ Biofilms Microbiomes 2024; 10:143. [PMID: 39632887 PMCID: PMC11618413 DOI: 10.1038/s41522-024-00619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Bacterial biofilms are highly adaptable and resilient to challenges. Nutrient availability can induce changes in biofilm growth, architecture and mechanical properties. Their extracellular matrix plays an important role in achieving biofilm stability under different environmental conditions. Curli amyloid fibers are critical for the architecture and stiffness of E. coli biofilms, but how this major matrix component adapts to different environmental cues remains unclear. We investigated, for the first time, the effect of nutrient availability both on biofilm material properties and on the structure and properties of curli amyloid fibers extracted from similar biofilms. Our results show that biofilms grown on low nutrient substrates are stiffer, contain more curli fibers, and these fibers present higher β-sheet content and chemical stability. Our multiscale study sheds new light on the relationship between bacterial matrix molecular structure and biofilm macroscopic properties. This knowledge will benefit the development of both anti-biofilm strategies and biofilm-based materials.
Collapse
Affiliation(s)
- Macarena Siri
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.
- Max Planck Queensland Centre, Potsdam, Germany.
| | - Mónica Vázquez-Dávila
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Carolina Sotelo Guzman
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Cécile M Bidan
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.
- Max Planck Queensland Centre, Potsdam, Germany.
| |
Collapse
|
11
|
Meyer N, Torrent J, Balme S. Characterizing Prion-Like Protein Aggregation: Emerging Nanopore-Based Approaches. SMALL METHODS 2024; 8:e2400058. [PMID: 38644684 PMCID: PMC11672191 DOI: 10.1002/smtd.202400058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/10/2024] [Indexed: 04/23/2024]
Abstract
Prion-like protein aggregation is characteristic of numerous neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. This process involves the formation of aggregates ranging from small and potentially neurotoxic oligomers to highly structured self-propagating amyloid fibrils. Various approaches are used to study protein aggregation, but they do not always provide continuous information on the polymorphic, transient, and heterogeneous species formed. This review provides an updated state-of-the-art approach to the detection and characterization of a wide range of protein aggregates using nanopore technology. For each type of nanopore, biological, solid-state polymer, and nanopipette, discuss the main achievements for the detection of protein aggregates as well as the significant contributions to the understanding of protein aggregation and diagnostics.
Collapse
Affiliation(s)
- Nathan Meyer
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonCedex 5Montpellier34095France
- INMUniversity of MontpellierINSERMMontpellier34095France
| | - Joan Torrent
- INMUniversity of MontpellierINSERMMontpellier34095France
| | - Sébastien Balme
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonCedex 5Montpellier34095France
| |
Collapse
|
12
|
Mohammad Karim A. Principles and Biomedical Applications of Self-Assembled Peptides: Potential Treatment of Type 2 Diabetes Mellitus. Pharmaceutics 2024; 16:1442. [PMID: 39598565 PMCID: PMC11597675 DOI: 10.3390/pharmaceutics16111442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is the most prevalent metabolic disorder worldwide. There have been tremendous efforts to find a safe and prolonged effective therapy for its treatment. Peptide hormones, from certain organisms in the human body, as the pharmaceutical agents, have shown outstanding profiles of efficacy and safety in plasma glucose regulation. Their therapeutic promises have undergone intensive investigations via examining their physicochemical and pharmacokinetic properties. Their major drawback is their short half-life in vivo. To address this challenge, researchers have recently started to apply the state-of-the-art molecular self-assembly on peptide hormones to form nanofibrillar structures, as a smart nanotherapeutic drug delivery technique, to tremendously enhance their prolonged bioactivity in vivo. This revolutionary therapeutic approach would significantly improve patient compliance. First, this review provides a comprehensive summary on the pathophysiology of T2DM, various efforts to treat this chronic disorder, and the limitations and drawbacks of these treatment approaches. Next, this review lays out detailed insights on various aspects of peptide self-assembly: adverse effects, potential applications in nanobiotechnology, thermodynamics and kinetics of the process, as well as the molecular structures of the self-assembled configurations. Furthermore, this review elucidates the recent efforts on applying reversible human-derived peptide self-assembly to generate highly organized smart nanostructured drug formulations known as nanofibrils to regulate and prolong the bioactivity of the human gut hormone peptides in vivo to treat T2DM. Finally, this review highlights the future research directions to advance the knowledge on the state-of-the-art peptide self-assembly process to apply the revolutionary smart nanotherapeutics for treatment of chronic disorders such as T2DM with highly improved patient compliance.
Collapse
Affiliation(s)
- Alireza Mohammad Karim
- Nanoscience Centre, Department of Engineering, University of Cambridge, 11 J. J. Thomson Avenue, Cambridge CB3 0FF, UK
| |
Collapse
|
13
|
Bilog M, Vedad J, Capadona C, Profit AA, Desamero RZB. Key charged residues influence the amyloidogenic propensity of the helix-1 region of serum amyloid A. Biochim Biophys Acta Gen Subj 2024; 1868:130690. [PMID: 39117048 PMCID: PMC11547331 DOI: 10.1016/j.bbagen.2024.130690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/15/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Increased plasma levels of serum amyloid A (SAA), an acute-phase protein that is secreted in response to inflammation, may lead to the accumulation of amyloid in various organs thereby obstructing their functions. Severe cases can lead to a systemic disorder called AA amyloidosis. Previous studies suggest that the N-terminal helix is the most amyloidogenic region of SAA. Moreover, computational studies implicated a significant role for Arg-1 and the residue-specific interactions formed during the fibrillization process. With a focus on the N-terminal region of helix-1, SAA1-13, mutational analysis was employed to interrogate the roles of the amino acid residues, Arg-1, Ser-5, Glu-9, and Asp-12. The truncated SAA1-13 fragment was systematically modified by substituting the key residues with alanine or uncharged but structurally similar amino acids. We monitored the changes in the amyloidogenic propensities, associated conformational markers, and morphology of the amyloids resulting from the mutation of SAA1-13. Mutating out Arg-1 resulted in much reduced aggregation propensity and a lack of detectable β-structures alluding to the importance of salt-bridge interactions involving Arg-1. Our data revealed that by systematically mutating the key amino acid residues, we can modulate the amyloidogenic propensity and alter the time-dependent conformational variation of the peptide. When the behaviors of each mutant peptide were analyzed, they provided evidence consistent with the aggregation pathway predicted by MD simulation studies. Here, we detail the important temporal molecular interactions formed by Arg-1 with Ser-5, Glu-9, and Asp-12 and discuss its mechanistic implications on the self-assembly of the helix-1 region of SAA.
Collapse
Affiliation(s)
- Marvin Bilog
- Department of Chemistry and the Institute of Macromolecular Assembly, York College of the City University of New York, Jamaica, New York 11451, United States; PhD Programs in Biochemistry, Graduate Center of the City University of New York, NY, New York, 10016, United States
| | - Jayson Vedad
- PhD Programs in Chemistry, Graduate Center of the City University of New York, NY, New York, 10016, United States; Chemistry and Biochemistry Department, Brooklyn College, 2900 Bedford Avenue, Brooklyn, New York, 11210, United States
| | - Charisse Capadona
- Department of Chemistry and the Institute of Macromolecular Assembly, York College of the City University of New York, Jamaica, New York 11451, United States
| | - Adam A Profit
- Department of Chemistry and the Institute of Macromolecular Assembly, York College of the City University of New York, Jamaica, New York 11451, United States; PhD Programs in Chemistry, Graduate Center of the City University of New York, NY, New York, 10016, United States; PhD Programs in Biochemistry, Graduate Center of the City University of New York, NY, New York, 10016, United States
| | - Ruel Z B Desamero
- Department of Chemistry and the Institute of Macromolecular Assembly, York College of the City University of New York, Jamaica, New York 11451, United States; PhD Programs in Chemistry, Graduate Center of the City University of New York, NY, New York, 10016, United States; PhD Programs in Biochemistry, Graduate Center of the City University of New York, NY, New York, 10016, United States.
| |
Collapse
|
14
|
Singh Y, Ahmad R, Raza A, Warsi MS, Mustafa M, Khan H, Hassan MI, Khan R, Moinuddin, Habib S. Exploring the effects of 4-chloro-o-phenylenediamine on human fibrinogen: A comprehensive investigation via biochemical, biophysical and computational approaches. Int J Biol Macromol 2024; 280:135825. [PMID: 39313050 DOI: 10.1016/j.ijbiomac.2024.135825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/28/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024]
Abstract
Fibrinogen (Fg), an essential plasma glycoprotein involved in the coagulation cascade, undergoes structural alterations upon exposure to various chemicals, impacting its functionality and contributing to pathological conditions. This research article explored the effects of 4-Chloro-o-phenylenediamine (4-Cl-o-PD), a common hair dye component (IUPAC = 1-Chloro-3,4-diaminobenzene), on human fibrinogen through comprehensive computational, biophysical, and biochemical approaches. The formation of a stable ligand-protein complex is confirmed through molecular docking and molecular dynamics simulations, revealing possible interaction having a favorable -4.8 kcal/mol binding energy. Biophysical results, including UV-vis and fluorescence spectroscopies, corroborated with the computational findings, whereas Fourier transform infrared spectroscopy (FT-IR) and circular dichroism spectroscopy (CD) provide insights into the alterations of secondary structures upon interaction with 4-Cl-o-PD. Anilinonaphthalene-sulfonic acid (ANS) fluorescence showed a partially unfolded protein, with enhanced α to β-sheet transition as evidenced by thioflavin T (ThT) spectroscopy and microscopy. Moreover, biochemical assays confirmed the formation of carbonyl compounds that may be responsible for the oxidation of methionine residues in fibrinogen. Electrophoresis and electron microscopy confirmed the formation of aggregates. Our findings elucidate the interaction pattern of 4-Cl-o-PD with Fg, leading to structural perturbation, which may have potential implications for fibrinogen misfolding or its aggregation. Protein aggregation or its misfolded products affect peripheral tissues and the central nervous system. Many chronic progressive diseases, like type II diabetes mellitus, Alzheimer's disease, Parkison's disease, and Creutzfeldt-Jakob disease are associated with intrinsically aberrant disordered proteins. Understanding these interactions may offer new perspectives on the safety and biocompatibility of dye compounds, which may contribute to developing improved strategies for acquired amyloidogenesis.
Collapse
Affiliation(s)
- Yogendra Singh
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Rizwan Ahmad
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Ali Raza
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Mohd Sharib Warsi
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Mohd Mustafa
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Hamda Khan
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ruhi Khan
- Department of Medicine, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Moinuddin
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Safia Habib
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India.
| |
Collapse
|
15
|
Kozina A, Herbert-Alonso G, Díaz A, Flores G, Guevara J. Effect of the aggregation state of amyloid-beta (25-35) on the brain oxidative stress in vivo. PLoS One 2024; 19:e0310258. [PMID: 39471144 PMCID: PMC11521274 DOI: 10.1371/journal.pone.0310258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/27/2024] [Indexed: 11/01/2024] Open
Abstract
Aggregation pathway of amyloid-β (25-35) in water affects the oxidative stress in the brain observed after administration of aggregated peptide in animals in vivo. Our studies on peptide aggregation ex situ prior to injection suggest that from the onset of peptide incubation in aqueous media, all samples exhibit the formation of fibril-like aggregates, characterized by a significant amount of β-sheets. This induces significant oxidative stress in vivo as observed for up to 60 min of peptide aggregation time. As the aggregation advances, the fibril-like aggregates become longer and intertwined, while the amount of β-sheets does not change significantly. An injection of such large, thick, and entangled aggregates in the animal brain results in a drastic increase in oxidative stress. This may be related to the number of activated microglia that initiate a sequence of inflammatory responses in the presence of large, highly interconnected fibrils.
Collapse
Affiliation(s)
- Anna Kozina
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Jorge Guevara
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
16
|
Kouhi ZH, Seyedalipour B, Hosseinkhani S, Chaichi MJ. Bisdemethoxycurcumin, a novel potent polyphenolic compound, effectively inhibits the formation of amyloid aggregates in ALS-associated hSOD1 mutant (L38R). Int J Biol Macromol 2024; 282:136701. [PMID: 39461630 DOI: 10.1016/j.ijbiomac.2024.136701] [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: 08/12/2024] [Revised: 09/29/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Protein misfolding is a biological process that leads to protein aggregation. Anomalous misfolding and aggregation of human superoxide dismutase (hSOD1) into amyloid aggregates is a characteristic feature of amyotrophic lateral sclerosis (ALS), a neurodegenerative illness. Thus, focusing on the L38R mutant may be a wise decision to comprehend the SOD1 disease process in ALS. We suggest that Bisdemethoxycurcumin (BDMC) may be a strong anti-amyloidogenic polyphenol against L38R mutant aggregation. Protein stability, hydrophobicity, and flexibility were altered when BDMC was bound to the L38R mutant, as shown by molecular dynamic (MD) simulations and molecular docking. FTIR data shows α-Helix dominance in BDMC-containing samples, with reduced β-sheet and disordered peaks, indicating the decrease of aggregate species. ThT aggregation kinetics curves show BDMC reduces L38R mutant aggregation dose-dependently, with higher BDMC concentrations yielding greater reductions. TEM images showed various quantities of amorphous aggregates, but notably, 60 μM BDMC markedly reduced aggregate density, underscoring BDMC's inhibitory effect. Hemolysis tests revealed aggregate species in BDMC-treated samples were less toxic than in L38R mutant samples alone at the same concentrations and exposure times. Overall, BDMC has substantial potential to develop highly effective inhibitors that mitigate the risk of fatal ALS.
Collapse
Affiliation(s)
- Zeinab Haghgoo Kouhi
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Bagher Seyedalipour
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran.
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javad Chaichi
- Department of Analytical Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
17
|
Giri A, Bhattacharya M. Intrinsic conformational preference in the monomeric protein governs amyloid polymorphism. Phys Chem Chem Phys 2024; 26:25222-25231. [PMID: 39315929 DOI: 10.1039/d4cp01973c] [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: 09/25/2024]
Abstract
The inherent stochasticity associated with the hierarchical self-assembly of either native-like or partially-unfolded protein monomers leads to the formation of transient, morphologically-diverse prefibrillar species resulting in structurally-distinct polymorphic protein aggregates. High-resolution structural characterization of mature aggregates has revealed heterogeneous supramolecular packing of protofibrils within amyloid polymorphs. However, little is known about whether initial monomeric protein conformers engender polymorphism at the onset of aggregation. Here, we show that intrinsic conformational preference in aggregation-competent monomeric ovalbumin, an archetypal serpin, dictates fibrillar polymorphism by modulating aggregation pathways. Using fluorescence, FT-IR, and vibrational Raman spectroscopy coupled with dynamic light scattering and electron microscopy, we demonstrate that conformationally-diverse amyloidogenic monomers, formed via an interplay of electrostatic and hydrophobic interactions before the commencement of aggregation, play a crucial role in promoting amyloid polymorphism. Moreover, the monomeric conformational fingerprints, accrued at the onset of aggregation, persist and propagate during the formation of polymorphic amyloids. Our results delineate essential conformational characteristics of the monomeric protein preceding aggregation, which will have broad implications in the mechanistic understanding of amyloid strain diversity observed in disease-related proteins.
Collapse
Affiliation(s)
- Anjali Giri
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala-147004, Punjab, India.
| | - Mily Bhattacharya
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala-147004, Punjab, India.
| |
Collapse
|
18
|
Tiroli-Cepeda AO, Linhares LA, Aragão AZB, de Jesus JR, Wasilewska-Sampaio AP, De Felice FG, Ferreira ST, Borges JC, Cyr DM, Ramos CHI. Type I Hsp40s/DnaJs aggregates exhibit features reminiscent of amyloidogenic structures. FEBS J 2024; 291:3904-3923. [PMID: 38975859 PMCID: PMC11468011 DOI: 10.1111/febs.17215] [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: 12/14/2023] [Revised: 03/14/2024] [Accepted: 06/20/2024] [Indexed: 07/09/2024]
Abstract
A rise in temperature triggers a structural change in the human Type I 40 kDa heat shock protein (Hsp40/DnaJ), known as DNAJA1. This change leads to a less compact structure, characterized by an increased presence of solvent-exposed hydrophobic patches and β-sheet-rich regions. This transformation is validated by circular dichroism, thioflavin T binding, and Bis-ANS assays. The formation of this β-sheet-rich conformation, which is amplified in the absence of zinc, leads to protein aggregation. This aggregation is induced not only by high temperatures but also by low ionic strength and high protein concentration. The aggregated conformation exhibits characteristics of an amyloidogenic structure, including a distinctive X-ray diffraction pattern, seeding competence (which stimulates the formation of amyloid-like aggregates), cytotoxicity, resistance to SDS, and fibril formation. Interestingly, the yeast Type I Ydj1 also tends to adopt a similar β-sheet-rich structure under comparable conditions, whereas Type II Hsp40s, whether human or from yeast, do not. Moreover, Ydj1 aggregates were found to be cytotoxic. Studies using DNAJA1- and Ydj1-deleted mutants suggest that the zinc-finger region plays a crucial role in amyloid formation. Our discovery of amyloid aggregation in a C-terminal deletion mutant of DNAJA1, which resembles a spliced homolog expressed in the testis, implies that Type I Hsp40 co-chaperones may generate amyloidogenic species in vivo.
Collapse
Affiliation(s)
- Ana O Tiroli-Cepeda
- Institute of Chemistry, Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| | - Leonardo A Linhares
- Institute of Chemistry, Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| | - Annelize Z B Aragão
- Institute of Chemistry, Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| | - Jemmyson R de Jesus
- Institute of Chemistry, Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| | | | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sérgio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Júlio C Borges
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, Brazil
| | | | - Carlos H I Ramos
- Institute of Chemistry, Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| |
Collapse
|
19
|
Guzzi R, Bartucci R. Thermal effects and drugs competition on the palmitate binding capacity of human serum albumin. Biochem Biophys Res Commun 2024; 722:150168. [PMID: 38797156 DOI: 10.1016/j.bbrc.2024.150168] [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: 03/14/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Human serum albumin (HSA) is the most abundant plasma protein of the circulatory system. It is a multidomain, multifunctional protein that, combining diverse affinities and wide specificity, binds, stores, and transports a variety of biological compounds, pharmacores, and fatty acids. HSA is finding increasing uses in drug-delivery due to its ability to carry functionalized ligands and prodrugs. All this raises the question of competition for binding sites occupancy in case of multiple ligands, which in turn influences the protein structure/dynamic/function relationship and also has an impact on the biomedical applications. In this work, the effects of interactive binding of palmitic acid (PA), warfarin (War) and ibuprofen (Ibu) on the thermal stability of HSA were studied using DSC, ATR-FTIR, and EPR. PA is a high-affinity physiological ligand, while the two drugs are widely used for their anticoagulant (War) and anti-inflammatory (Ibu) efficacy, and are exogenous compounds that accommodate in the deputed drug site DS1 and DS2, respectively overlapping with some of the fatty acid binding sites. The results indicate that HSA acquires the highest thermal stability when it is fully saturated with PA. The binding of this physiological ligand does not hamper the binding of War or Ibu to the native state of the protein. In addition, the three ligands bind simultaneously, suggesting a synergic cooperative influence due to allosteric effects. The increased thermal stability subsequent to binary and multiple ligands binding moderates protein aggregation propensity and restricts protein dynamics. The biophysics findings provide interesting features about protein stability, aggregation, and dynamics in interaction with multiple ligands and are relevant in drug-delivery.
Collapse
Affiliation(s)
- Rita Guzzi
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036, Rende, Italy; CNR-NANOTEC, Department of Physics, University of Calabria, 87036, Rende, Italy.
| | - Rosa Bartucci
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036, Rende, Italy
| |
Collapse
|
20
|
Pei R, Tan J, Luo Y, Ye S. Close Packing in Trans Conformers Promotes the Formation of Supramolecular Structures with C 1 Symmetry. J Phys Chem Lett 2024; 15:8797-8803. [PMID: 39166774 DOI: 10.1021/acs.jpclett.4c01857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Assemblies with C1 symmetry exhibit important applications in many fields such as enantioselective catalysis. However, their formation is challenging due to their large entropic disadvantage, and molecular information on their formation dynamics is limited because of the lack of effective characterization techniques. Here, using achiral amphiphilic molecules such as N-oleoyl ethanolamide (OEA) and its analogues as modeling assembly units, we demonstrated that the sss polarization signals, generated by femtosecond sum frequency generation vibrational spectroscopy (SFG-VS), provide a powerful tool to monitor the formation dynamics of the C1 symmetric supramolecular structures at the interfaces. The trans conformation of the assembly units can provide strong π-π interactions and thus produce enough enthalpy to drive the formation of C1 symmetric supramolecular structures. However, the cis conformation impedes the assembly of C1 symmetric structures and cannot generate sss and chiral polarization SFG signals. These findings may aid in rationally constructing ordered and functional superstructures and understanding the mechanism of chirality formation.
Collapse
Affiliation(s)
- Ruoqi Pei
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Junjun Tan
- Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China
| | - Yi Luo
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China
| | - Shuji Ye
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China
| |
Collapse
|
21
|
Guo Z, Chiesa G, Yin J, Sanford A, Meier S, Khalil AS, Cheng JX. Structural Mapping of Protein Aggregates in Live Cells Modeling Huntington's Disease. Angew Chem Int Ed Engl 2024; 63:e202408163. [PMID: 38880765 PMCID: PMC11781839 DOI: 10.1002/anie.202408163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
While protein aggregation is a hallmark of many neurodegenerative diseases, acquiring structural information on protein aggregates inside live cells remains challenging. Traditional microscopy does not provide structural information on protein systems. Routinely used fluorescent protein tags, such as Green Fluorescent Protein (GFP), might perturb native structures. Here, we report a counter-propagating mid-infrared photothermal imaging approach enabling mapping of secondary structure of protein aggregates in live cells modeling Huntington's disease. By comparing mid-infrared photothermal spectra of label-free and GFP-tagged huntingtin inclusions, we demonstrate that GFP fusions indeed perturb the secondary structure of aggregates. By implementing spectra with small spatial step for dissecting spectral features within sub-micrometer distances, we reveal that huntingtin inclusions partition into a β-sheet-rich core and a ɑ-helix-rich shell. We further demonstrate that this structural partition exists only in cells with the [RNQ+] prion state, while [rnq-] cells only carry smaller β-rich non-toxic aggregates. Collectively, our methodology has the potential to unveil detailed structural information on protein assemblies in live cells, enabling high-throughput structural screenings of macromolecular assemblies.
Collapse
Affiliation(s)
- Zhongyue Guo
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Photonics Center, Boston University, Boston, MA 02215, USA
| | - Giulio Chiesa
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Jiaze Yin
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| | - Adam Sanford
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Stefan Meier
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Ahmad S Khalil
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Ji-Xin Cheng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
22
|
Kim SM, Chang YR, Melby J, Kim YJ, Davis D, Lee YJ. Quantum Cascade Laser Infrared Spectroscopy for Glycan Analysis of Glycoprotein Solutions. Anal Chem 2024; 96:13120-13130. [PMID: 39078866 DOI: 10.1021/acs.analchem.4c01772] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Glycans are oligosaccharides attached to proteins or lipids and affect their functions, such as drug efficacy, structural contribution, metabolism, immunogenicity, and molecular recognition. Conventional glycosylation analysis has relied on destructive, slow, system-sensitive methods, including enzymatic reactions, chromatography, fluorescence labeling, and mass spectrometry. Herein, we propose quantum cascade laser (QCL) infrared (IR) spectroscopy as a rapid, nondestructive method to quantify glycans and their monosaccharide composition. Previously, we demonstrated high-sensitivity IR spectroscopy of protein solution using solvent absorption compensation (SAC) and double-beam modulation (DBM) techniques. However, the SAC-DBM approach suffered a limited frequency scanning range (<400 cm-1) due to the light dispersion by acousto-optic modulators (AOMs). Here, we implemented a mirror-based double-pass AOM in the SAC-DBM scheme and successfully extended the frequency range to (970 to 1840 cm-1), which encompasses the vibrational fingerprint of biomolecules. The extended frequency range allowed the simultaneous observation of monosaccharide ring bands (1000 to 1200 cm-1) and protein amide bands (1500 to 1700 cm-1). We compared the IR spectra of six glycoproteins and two nonglycosylated proteins with the results from intact mass spectrometry. The IR absorbance ratios of the ring band to the amide band of glycoproteins in solutions showed a linear correlation with the ratios of glycan to protein backbone masses. Furthermore, a multivariate analysis produced monosaccharide compositions consistent with the reported database for the glycoproteins, and the monosaccharide compositions were used to improve the predictability of the glycan-protein mass ratio from the IR-absorbance ratio. This nondestructive, high-sensitivity QCL-IR spectroscopy could be used as a standard method to monitor batch-to-batch comparability during drug manufacturing and quantify the glycosylation and monosaccharide composition of new glycoproteins and other glycosylated biosystems.
Collapse
Affiliation(s)
- Seong-Min Kim
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Yow-Ren Chang
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Jake Melby
- Process and Analytical Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Yoen Joo Kim
- Process and Analytical Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Darryl Davis
- Process and Analytical Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Young Jong Lee
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
23
|
Dürvanger Z, Bencs F, Menyhárd DK, Horváth D, Perczel A. Solvent induced amyloid polymorphism and the uncovering of the elusive class 3 amyloid topology. Commun Biol 2024; 7:968. [PMID: 39122990 PMCID: PMC11316126 DOI: 10.1038/s42003-024-06621-8] [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: 12/01/2023] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Aggregation-prone-motifs (APRs) of proteins are short segments, which - as isolated peptides - form diverse amyloid-like crystals. We introduce two APRs - designed variants of the incretin mimetic Exendin-4 - that both display crystal-phase polymorphism. Crystallographic and spectroscopic analysis revealed that a single amino-acid substitution can greatly reduce topological variability: while LYIQWL can form both parallel and anti-parallel β-sheets, LYIQNL selects only the former. We also found that the parallel/anti-parallel switch of LYIQWL can be induced by simply changing the crystallization temperature. One crystal form of LYIQNL was found to belong to the class 3 topology, an arrangement previously not encountered among proteinogenic systems. We also show that subtle environmental changes lead to crystalline assemblies with different topologies, but similar interfaces. Spectroscopic measurements showed that polymorphism is already apparent in the solution state. Our results suggest that the temperature-, sequence- and environmental sensitivity of physiological amyloids is reflected in assemblies of the APR segments, which, complete with the new class 3 crystal form, effectively sample all the originally proposed basic topologies of amyloid-like aggregates.
Collapse
Affiliation(s)
- Zsolt Dürvanger
- Laboratory of Structural Chemistry and Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- HUN-REN-ELTE Protein Modeling Research Group, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
| | - Fruzsina Bencs
- Laboratory of Structural Chemistry and Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- Hevesy György PhD School of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
| | - Dóra K Menyhárd
- Laboratory of Structural Chemistry and Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- HUN-REN-ELTE Protein Modeling Research Group, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
| | - Dániel Horváth
- Laboratory of Structural Chemistry and Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- HUN-REN-ELTE Protein Modeling Research Group, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
| | - András Perczel
- Laboratory of Structural Chemistry and Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary.
- HUN-REN-ELTE Protein Modeling Research Group, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary.
| |
Collapse
|
24
|
Sarma S, Sudarshan TR, Nguyen V, Robang AS, Xiao X, Le JV, Helmicki ME, Paravastu AK, Hall CK. Design of parallel 𝛽-sheet nanofibrils using Monte Carlo search, coarse-grained simulations, and experimental testing. Protein Sci 2024; 33:e5102. [PMID: 39037281 PMCID: PMC11261811 DOI: 10.1002/pro.5102] [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: 11/10/2023] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/23/2024]
Abstract
Peptide self-assembly into amyloid fibrils provides numerous applications in drug delivery and biomedical engineering applications. We augment our previously-established computational screening technique along with experimental biophysical characterization to discover 7-mer peptides that self-assemble into "parallel β-sheets", that is, β-sheets with N-terminus-to-C-terminus 𝛽-strand vectors oriented in parallel. To accomplish the desired β-strand organization, we applied the PepAD amino acid sequence design software to the Class-1 cross-β spine defined by Sawaya et al. This molecular configuration includes two layers of parallel β-sheets stacked such that N-terminus-to-C-terminus vectors are oriented antiparallel for molecules on adjacent β-sheets. The first cohort of PepAD identified peptides were examined for their fibrillation behavior in DMD/PRIME20 simulations, and the top performing sequence was selected as a prototype for a subsequent round of sequence refinement. The two rounds of design resulted in a library of eight 7-mer peptides. In DMD/PRIME20 simulations, five of these peptides spontaneously formed fibril-like structures with a predominantly parallel 𝛽-sheet arrangement, two formed fibril-like structure with <50% in parallel 𝛽-sheet arrangement and one remained a random coil. Among the eight candidate peptides produced by PepAD and DMD/PRIME20, five were synthesized and purified. All five assembled into amyloid fibrils composed of parallel β-sheets based on Fourier transform infrared spectroscopy, circular dichroism, electron microscopy, and thioflavin-T fluorescence spectroscopy measurements.
Collapse
Affiliation(s)
- Sudeep Sarma
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Tarunya Rao Sudarshan
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Van Nguyen
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Alicia S. Robang
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Xingqing Xiao
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
- Present address:
Department of Chemistry, School of Chemistry and Chemical EngineeringHainan UniversityHaikou CityHainan ProvincePeople's Republic of China
| | - Justin V. Le
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Michael E. Helmicki
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Anant K. Paravastu
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Carol K. Hall
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
| |
Collapse
|
25
|
Espargaró A, Álvarez-Berbel I, Busquets MA, Sabate R. In Vivo Assays for Amyloid-Related Diseases. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:433-458. [PMID: 38598824 DOI: 10.1146/annurev-anchem-061622-023326] [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: 04/12/2024]
Abstract
Amyloid-related diseases, such as Alzheimer's and Parkinson's disease, are devastating conditions caused by the accumulation of abnormal protein aggregates known as amyloid fibrils. While assays involving animal models are essential for understanding the pathogenesis and developing therapies, a wide array of standard analytical techniques exists to enhance our understanding of these disorders. These techniques provide valuable information on the formation and propagation of amyloid fibrils, as well as the pharmacokinetics and pharmacodynamics of candidate drugs. Despite ethical concerns surrounding animal use, animal models remain vital tools in the search for treatments. Regardless of the specific animal model chosen, the analytical methods used are usually standardized. Therefore, the main objective of this review is to categorize and outline the primary analytical methods used in in vivo assays for amyloid-related diseases, highlighting their critical role in furthering our understanding of these disorders and developing effective therapies.
Collapse
Affiliation(s)
- Alba Espargaró
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Irene Álvarez-Berbel
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
| | - Maria Antònia Busquets
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sabate
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Dey A, Patil A, Arumugam S, Maiti S. Single-Molecule Maps of Membrane Insertion by Amyloid-β Oligomers Predict Their Toxicity. J Phys Chem Lett 2024; 15:6292-6298. [PMID: 38855822 DOI: 10.1021/acs.jpclett.4c01048] [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: 06/11/2024]
Abstract
The interaction of small Amyloid-β (Aβ) oligomers with the lipid membrane is an important component of the pathomechanism of Alzheimer's disease (AD). However, oligomers are heterogeneous in size. How each type of oligomer incorporates into the membrane, and how that relates to their toxicity, is unknown. Here, we employ a single molecule technique called Q-SLIP (Quencher-induced Step Length Increase in Photobleaching) to measure the membrane insertion of each monomeric unit of individual oligomers of Aβ42, Aβ40, and Aβ40-F19-Cyclohexyl alanine (Aβ40-F19Cha), and correlate it with their toxicity. We observe that the N-terminus of Aβ42 inserts close to the center of the bilayer, the less toxic Aβ40 inserts to a shallower depth, and the least toxic Aβ40-F19Cha has no specific distribution. This oligomer-specific map provides a mechanistic representation of membrane-mediated Aβ toxicity and should be a valuable tool for AD research.
Collapse
Affiliation(s)
- Arpan Dey
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Abhishek Patil
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC 3800, Australia
| | - Senthil Arumugam
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC 3800, Australia
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, VIC 3800, Australia
| | - Sudipta Maiti
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| |
Collapse
|
27
|
Matsui M, Murata T, Kurobe-Takashima Y, Ikeda T, Noguchi-Shinohara M, Ono K, Shidara H, Otsuka K, Kuriki D, Suzuki M, Kobayashi S. Lutein from Chicken Eggs Prevents Amyloid β-Peptide Aggregation In Vitro and Amyloid β-Induced Inflammation in Human Macrophages (THP-1). ACS OMEGA 2024; 9:26616-26627. [PMID: 38911805 PMCID: PMC11191573 DOI: 10.1021/acsomega.4c03353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 06/25/2024]
Abstract
Epidemiological studies predict that chicken eggs contain constituents other than proteins that prevent Alzheimer's disease. This study screened for constituents that inhibit the aggregation of amyloid β peptide (Aβ)1-42 and elucidated their mechanisms to explore the active components of chicken eggs. Thioflavin T assays and transmission electron microscopy observations showed that arachidonic acid (ARA), lysophosphatidylcholine, lutein (LTN), palmitoleic acid, and zeaxanthin inhibited Aβ aggregation. Among these, ARA and LTN showed the highest activity. Photoinduced cross-linking of unmodified protein assays and infrared absorption spectrometry measurements showed that LTN strongly inhibited highly toxic Aβ1-42 protofibril formation. Furthermore, LTN suppressed Aβ1-42-induced IL 1B and TNF expression in human macrophage-like cells. In summary, LTN plays a crucial role in the AD-preventive effect of chicken eggs by suppressing Aβ1-42 aggregation and Aβ1-42-induced inflammation.
Collapse
Affiliation(s)
- Misuzu Matsui
- Department
of Applied Biological Chemistry, Graduate School of Agricultural and
Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Tomofusa Murata
- Department
of Applied Biological Chemistry, Graduate School of Agricultural and
Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Yuki Kurobe-Takashima
- Department
of Applied Biological Chemistry, Graduate School of Agricultural and
Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Tokuhei Ikeda
- Department
of Neurology, Kanazawa University Graduate
School of Medical Sciences, Kanazawa 920-1192, Japan
| | - Moeko Noguchi-Shinohara
- Department
of Neurology, Kanazawa University Graduate
School of Medical Sciences, Kanazawa 920-1192, Japan
| | - Kenjiro Ono
- Department
of Neurology, Kanazawa University Graduate
School of Medical Sciences, Kanazawa 920-1192, Japan
| | | | - Kurataka Otsuka
- R&D
Division, Kewpie Corporation, Tokyo 150-0002, Japan
- Division
of Translational Oncology, Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Tokyo
NODAI Research Institute, Tokyo University
of Agriculture, Tokyo 156-8502, Japan
- Division
of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Daisuke Kuriki
- R&D
Division, Kewpie Corporation, Tokyo 150-0002, Japan
- Division
of Translational Oncology, Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Michio Suzuki
- Department
of Applied Biological Chemistry, Graduate School of Agricultural and
Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Shoko Kobayashi
- Department
of Applied Biological Chemistry, Graduate School of Agricultural and
Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| |
Collapse
|
28
|
Robang A, Wong KM, Leisen J, Liu R, Radford WL, Rao Sudarshan T, Hudalla GA, Paravastu AK. Parallel β-Sheet Structure and Structural Heterogeneity Detected within Q11 Self-Assembling Peptide Nanofibers. J Phys Chem B 2024; 128:5387-5396. [PMID: 38787393 PMCID: PMC11163420 DOI: 10.1021/acs.jpcb.4c00825] [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: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Q11 peptide nanofibers are used as a biomaterial for applications such as antigen presentation and tissue engineering, yet detailed knowledge of molecular-level structure has not been reported. The Q11 peptide sequence was designed using heuristics-based patterning of hydrophobic and polar amino acids with oppositely charged amino acids placed at opposite ends of the sequence to promote antiparallel β-sheet formation. In this work, we employed solid-state nuclear magnetic resonance spectroscopy (NMR) to evaluate whether the molecular organization within Q11 self-assembled peptide nanofibers is consistent with the expectations of the peptide designers. We discovered that Q11 forms a distribution of molecular structures. NMR data from two-dimensional (2D) 13C-13C dipolar-assisted rotational resonance indicate that the K3 and E9 residues between Q11 β-strands are spatially proximate (within ∼0.6 nm). Frequency-selective rotational echo double resonance (fsREDOR) on K3 Nζ and E9 Cδ-labeled sites showed that approximately 9% of the sites are close enough for salt bridge formation to occur. Surprisingly, dipolar recoupling measurements revealed that Q11 peptides do not assemble into antiparallel β-sheets as expected, and structural analysis using Fourier-transform infrared spectroscopy and 2D NMR alone can be misleading. 13C PITHIRDS-CT dipolar recoupling measurements showed that the most abundant structure consists of parallel β-sheets, in contrast to the expected antiparallel β-sheet structure. Structural heterogeneity was detected from 15N{13C} REDOR measurements, with approximately 22% of β-strands having antiparallel nearest neighbors. We cannot propose a complete structural model of Q11 nanofibers because of the complexity involved when examining structurally heterogeneous samples using NMR. Altogether, our results show that while heuristics-based patterning is effective in promoting β-sheet formation, designing a peptide sequence to form a targeted β-strand arrangement remains challenging.
Collapse
Affiliation(s)
- Alicia
S. Robang
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Kong M. Wong
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Johannes Leisen
- School
of Chemistry & Biochemistry, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| | - Renjie Liu
- J. Crayton
Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Walker L. Radford
- School
of Chemistry & Biochemistry, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| | - Tarunya Rao Sudarshan
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Gregory A. Hudalla
- J. Crayton
Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Anant K. Paravastu
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
29
|
Wang Y, Liang X, Andrikopoulos N, Tang H, He F, Yin X, Li Y, Ding F, Peng G, Mortimer M, Ke PC. Remediation of Metal Oxide Nanotoxicity with a Functional Amyloid. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310314. [PMID: 38582521 PMCID: PMC11187920 DOI: 10.1002/advs.202310314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/21/2024] [Indexed: 04/08/2024]
Abstract
Understanding the environmental health and safety of nanomaterials (NanoEHS) is essential for the sustained development of nanotechnology. Although extensive research over the past two decades has elucidated the phenomena, mechanisms, and implications of nanomaterials in cellular and organismal models, the active remediation of the adverse biological and environmental effects of nanomaterials remains largely unexplored. Inspired by recent developments in functional amyloids for biomedical and environmental engineering, this work shows their new utility as metallothionein mimics in the strategically important area of NanoEHS. Specifically, metal ions released from CuO and ZnO nanoparticles are sequestered through cysteine coordination and electrostatic interactions with beta-lactoglobulin (bLg) amyloid, as revealed by inductively coupled plasma mass spectrometry and molecular dynamics simulations. The toxicity of the metal oxide nanoparticles is subsequently mitigated by functional amyloids, as validated by cell viability and apoptosis assays in vitro and murine survival and biomarker assays in vivo. As bLg amyloid fibrils can be readily produced from whey in large quantities at a low cost, the study offers a crucial strategy for remediating the biological and environmental footprints of transition metal oxide nanomaterials.
Collapse
Affiliation(s)
- Yue Wang
- School of Biomedical Sciences and EngineeringGuangzhou International CampusSouth China University of TechnologyGuangzhou510006China
- Nanomedicine CenterGreat Bay Area National Institute for Nanotechnology Innovation136 Kaiyuan AvenueGuangzhou510700China
| | - Xiufang Liang
- School of Biomedical Sciences and EngineeringGuangzhou International CampusSouth China University of TechnologyGuangzhou510006China
- Nanomedicine CenterGreat Bay Area National Institute for Nanotechnology Innovation136 Kaiyuan AvenueGuangzhou510700China
| | - Nicholas Andrikopoulos
- Nanomedicine CenterGreat Bay Area National Institute for Nanotechnology Innovation136 Kaiyuan AvenueGuangzhou510700China
- Drug DeliveryDisposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| | - Huayuan Tang
- Department of Engineering MechanicsHohai UniversityNanjing211100China
- Department of Physics and AstronomyClemson UniversityClemsonSC29634USA
| | - Fei He
- College of Environmental Science and EngineeringKey Laboratory of Yangtze River Water EnvironmentTongji University1239 Siping RoadShanghai200092China
| | - Xiang Yin
- College of Environmental Science and EngineeringKey Laboratory of Yangtze River Water EnvironmentTongji University1239 Siping RoadShanghai200092China
| | - Yuhuan Li
- Drug DeliveryDisposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Liver Cancer InstituteZhongshan HospitalKey Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationFudan UniversityShanghai200032China
| | - Feng Ding
- Department of Physics and AstronomyClemson UniversityClemsonSC29634USA
| | - Guotao Peng
- College of Environmental Science and EngineeringKey Laboratory of Yangtze River Water EnvironmentTongji University1239 Siping RoadShanghai200092China
| | - Monika Mortimer
- Laboratory of Environmental ToxicologyNational Institute of Chemical Physics and BiophysicsAkadeemia tee 23Tallinn12618Estonia
| | - Pu Chun Ke
- Nanomedicine CenterGreat Bay Area National Institute for Nanotechnology Innovation136 Kaiyuan AvenueGuangzhou510700China
- Drug DeliveryDisposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| |
Collapse
|
30
|
Nešić MD, Popović IA, Žakula J, Korićanac L, Filipović Tričković J, Valenta Šobot A, Jiménez MV, Algarra M, Dučić T, Stepić M. Synergistic Enhancement of Targeted Wound Healing by Near-Infrared Photodynamic Therapy and Silver Metal-Organic Frameworks Combined with S- or N-Doped Carbon Dots. Pharmaceutics 2024; 16:671. [PMID: 38794333 PMCID: PMC11124918 DOI: 10.3390/pharmaceutics16050671] [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: 03/11/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
The literature data emphasize that nanoparticles might improve the beneficial effects of near-infrared light (NIR) on wound healing. This study investigates the mechanisms of the synergistic wound healing potential of NIR light and silver metal-organic frameworks combined with nitrogen- and sulfur-doped carbon dots (AgMOFsN-CDs and AgMOFsS-CDs, respectively), which was conducted by testing the fibroblasts viability, scratch assays, biochemical analysis, and synchrotron-based Fourier transform infrared (SR-FTIR) cell spectroscopy and imaging. Our findings reveal that the combined treatment of AgMOFsN-CDs and NIR light significantly increases cell viability to nearly 150% and promotes cell proliferation, with reduced interleukin-1 levels, suggesting an anti-inflammatory response. SR-FTIR spectroscopy shows this combined treatment results in unique protein alterations, including increased α-helix structures and reduced cross-β. Additionally, protein synthesis was enhanced upon the combined treatment. The likely mechanism behind the observed changes is the charge-specific interaction of N-CDs from the AgMOFsN-CDs with proteins, enhanced by NIR light due to the nanocomposite's optical characteristics. Remarkably, the complete wound closure in the in vitro scratch assay was achieved exclusively with the combined NIR and AgMOFsN-CDs treatment, demonstrating the promising application of combined AgMOFsN-CDs with NIR light photodynamic therapy in regenerative nanomedicine and tissue engineering.
Collapse
Affiliation(s)
- Maja D. Nešić
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Iva A. Popović
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jelena Žakula
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.Ž.); (L.K.)
| | - Lela Korićanac
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.Ž.); (L.K.)
| | - Jelena Filipović Tričković
- Department of Physical Chemistry, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.F.T.); (A.V.Š.)
| | - Ana Valenta Šobot
- Department of Physical Chemistry, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (J.F.T.); (A.V.Š.)
| | | | - Manuel Algarra
- Department of Science, INAMAT—Institute for Advanced Materials and Mathematics, Public University of Navarra, 31006 Pamplona, Spain;
| | - Tanja Dučić
- MIRAS Beamline, ALBA-CELLS Synchrotron, 08290 Cerdanyola del Vallès, Spain;
| | - Milutin Stepić
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
31
|
Juković M, Ratkaj I, Kalafatovic D, Bradshaw NJ. Amyloids, amorphous aggregates and assemblies of peptides - Assessing aggregation. Biophys Chem 2024; 308:107202. [PMID: 38382283 DOI: 10.1016/j.bpc.2024.107202] [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: 11/29/2023] [Revised: 01/31/2024] [Accepted: 02/14/2024] [Indexed: 02/23/2024]
Abstract
Amyloid and amorphous aggregates represent the two major categories of aggregates associated with diseases, and although exhibiting distinct features, researchers often treat them as equivalent, which demonstrates the need for more thorough characterization. Here, we compare amyloid and amorphous aggregates based on their biochemical properties, kinetics, and morphological features. To further decipher this issue, we propose the use of peptide self-assemblies as minimalistic models for understanding the aggregation process. Peptide building blocks are significantly smaller than proteins that participate in aggregation, however, they make a plausible means to bridge the gap in discerning the aggregation process at the more complex, protein level. Additionally, we explore the potential use of peptide-inspired models to research the liquid-liquid phase separation as a feasible mechanism preceding amyloid formation. Connecting these concepts can help clarify our understanding of aggregation-related disorders and potentially provide novel drug targets to impede and reverse these serious illnesses.
Collapse
Affiliation(s)
- Maja Juković
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia
| | - Ivana Ratkaj
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia
| | - Daniela Kalafatovic
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia.
| | - Nicholas J Bradshaw
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia.
| |
Collapse
|
32
|
Daou D, Zarate Y, Maaloum M, Collin D, Fleith G, Constantin D, Moulin E, Giuseppone N. Out-of-Equilibrium Mechanical Disruption of β-Amyloid-Like Fibers using Light-Driven Molecular Motors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311293. [PMID: 38236822 DOI: 10.1002/adma.202311293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/10/2024] [Indexed: 01/26/2024]
Abstract
Artificial molecular motors have the potential to generate mechanical work on their environment by producing autonomous unidirectional motions when supplied with a source of energy. However, the harnessing of this mechanical work to subsequently activate various endoenergetic processes that can be useful in materials science remains elusive. Here, it is shown that by integrating a light-driven rotary motor through hydrogen bonds in a β-amyloid-like structure forming supramolecular hydrogels, the mechanical work generated during the constant rotation of the molecular machine under UV irradiation is sufficient to disrupt the β-amyloid fibers and to trigger a gel-to-sol transition at macroscopic scale. This melting of the gel under UV irradiation occurs 25 °C below the temperature needed to melt it by solely using thermal activation. In the dark, a reversible sol-gel transition is observed as the system fully recovers its original microstructure, thus illustrating the possible access to new kinds of motorized materials that can be controlled by advanced out-of-equilibrium thermodynamics.
Collapse
Affiliation(s)
- Dania Daou
- SAMS Research Group, CNRS, Université de Strasbourg, Institut Charles Sadron UPR 22, Strasbourg, 67000, France
| | - Yohan Zarate
- SAMS Research Group, CNRS, Université de Strasbourg, Institut Charles Sadron UPR 22, Strasbourg, 67000, France
| | - Mounir Maaloum
- SAMS Research Group, CNRS, Université de Strasbourg, Institut Charles Sadron UPR 22, Strasbourg, 67000, France
| | | | | | - Doru Constantin
- CNRS, Institut Charles Sadron UPR 22, Strasbourg, 67000, France
| | - Emilie Moulin
- SAMS Research Group, CNRS, Université de Strasbourg, Institut Charles Sadron UPR 22, Strasbourg, 67000, France
| | - Nicolas Giuseppone
- SAMS Research Group, CNRS, Université de Strasbourg, Institut Charles Sadron UPR 22, Strasbourg, 67000, France
- Institut Universitaire de France (IUF), Paris, 75005, France
| |
Collapse
|
33
|
Maity B, Kameyama S, Tian J, Pham TT, Abe S, Chatani E, Murata K, Ueno T. Fusion of amyloid beta with ferritin yields an isolated oligomeric beta-sheet-rich aggregate inside the ferritin cage. Biomater Sci 2024; 12:2408-2417. [PMID: 38511491 DOI: 10.1039/d4bm00173g] [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: 03/22/2024]
Abstract
Alzheimer's disease is a severe brain condition caused by the formation of amyloid plaques composed of amyloid beta (Aβ) peptides. These peptides form oligomers, protofibrils, and fibrils before deposition into amyloid plaques. Among these intermediates, Aβ oligomers (AβOs) were found to be the most toxic and therefore an appealing target for drug development and understanding their role in the disease. However, precise isolation and characterization of AβOs have proven challenging because AβOs tend to aggregate and form heterogeneous mixtures in solution. As a solution, we genetically fused the Aβ peptide with a ferritin monomer. Such fusion allowed the encapsulation of precisely 24 Aβ peptides inside the 24-mer ferritin cage. Using high-speed atomic force microscopy (HS-AFM), we disassembled ferritin and directly visualized the Aβ core enclosed within the cage. The thioflavin-T assay (ThT) and attenuated total reflection infrared spectroscopy (ATR-IR) revealed the presence of a β-sheet structure in the encapsulated oligomeric aggregate. Gallic acid, an amyloid inhibitor, can inhibit the fluorescence of ThT bound AβOs. Our approach represents a significant advancement in the isolation and characterization of β-sheet rich AβOs and is expected to be useful for future studies of other disordered peptides such as α-synuclein and tau.
Collapse
Affiliation(s)
- Basudev Maity
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Shiori Kameyama
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Jiaxin Tian
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Thuc Toan Pham
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Satoshi Abe
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Eri Chatani
- Department of Chemistry, Graduate School of Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Kazuyoshi Murata
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institute for Natural Sciences, Okazaki, Aichi, 444-8585, Japan
- National Institute for Physiological Sciences (NIPS), National Institute for Natural Sciences, Okazaki, Aichi, 444-8585, Japan
| | - Takafumi Ueno
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
- Living Systems Materialogy (LiSM) Research Group, International Research Frontiers Initiative (IRFI), Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
34
|
Nazarova A, Shiabiev I, Shibaeva K, Mostovaya O, Mukhametzyanov T, Khannanov A, Evtugyn V, Zelenikhin P, Shi X, Shen M, Padnya P, Stoikov I. Thiacalixarene Carboxylic Acid Derivatives as Inhibitors of Lysozyme Fibrillation. Int J Mol Sci 2024; 25:4721. [PMID: 38731940 PMCID: PMC11083589 DOI: 10.3390/ijms25094721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Amyloid fibroproliferation leads to organ damage and is associated with a number of neurodegenerative diseases affecting populations worldwide. There are several ways to protect against fibril formation, including inhibition. A variety of organic compounds based on molecular recognition of amino acids within the protein have been proposed for the design of such inhibitors. However, the role of macrocyclic compounds, i.e., thiacalix[4]arenes, in inhibiting fibrillation is still almost unknown. In the present work, the use of water-soluble thiacalix[4]arene derivatives for the inhibition of hen egg-white lysozyme (HEWL) amyloid fibrillation is proposed for the first time. The binding of HEWL by the synthesized thiacalix[4]arenes (logKa = 5.05-5.13, 1:1 stoichiometry) leads to the formation of stable supramolecular systems capable of stabilizing the protein structure and protecting against fibrillation by 29-45%. The macrocycle conformation has little effect on protein binding strength, and the native HEWL secondary structure does not change via interaction. The synthesized compounds are non-toxic to the A549 cell line in the range of 0.5-250 µg/mL. The results obtained may be useful for further investigation of the anti-amyloidogenic role of thiacalix[4]arenes, and also open up future prospects for the creation of new ways to prevent neurodegenerative diseases.
Collapse
Affiliation(s)
- Anastasia Nazarova
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Igor Shiabiev
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Ksenia Shibaeva
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Olga Mostovaya
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Timur Mukhametzyanov
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Arthur Khannanov
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Vladimir Evtugyn
- Interdisciplinary Center of Analytical Microscopy, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Pavel Zelenikhin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- CQM—Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Pavel Padnya
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Ivan Stoikov
- A. M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| |
Collapse
|
35
|
Zhao X, Mou C, Xu J, Cui W, Shi Y, Wang Y, Luo T, Guo W, Ye J, Chen W. Protection of Si Nanowires against A β Toxicity by the Inhibition of A β Aggregation. Molecules 2024; 29:1980. [PMID: 38731472 PMCID: PMC11085270 DOI: 10.3390/molecules29091980] [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: 03/26/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid beta (Aβ) plaques in the brain. Aβ1-42 is the main component of Aβ plaque, which is toxic to neuronal cells. Si nanowires (Si NWs) have the advantages of small particle size, high specific surface area, and good biocompatibility, and have potential application prospects in suppressing Aβ aggregation. In this study, we employed the vapor-liquid-solid (VLS) growth mechanism to grow Si NWs using Au nanoparticles as catalysts in a plasma-enhanced chemical vapor deposition (PECVD) system. Subsequently, these Si NWs were transferred to a phosphoric acid buffer solution (PBS). We found that Si NWs significantly reduced cell death in PC12 cells (rat adrenal pheochromocytoma cells) induced by Aβ1-42 oligomers via double staining with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and fluorescein diacetate/propyl iodide (FDA/PI). Most importantly, pre-incubated Si NWs largely prevented Aβ1-42 oligomer-induced PC12 cell death, suggesting that Si NWs exerts an anti-Aβ neuroprotective effect by inhibiting Aβ aggregation. The analysis of Fourier Transform Infrared (FTIR) results demonstrates that Si NWs reduce the toxicity of fibrils and oligomers by intervening in the formation of β-sheet structures, thereby protecting the viability of nerve cells. Our findings suggest that Si NWs may be a potential therapeutic agent for AD by protecting neuronal cells from the toxicity of Aβ1-42.
Collapse
Affiliation(s)
- Xuechun Zhao
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| | - Chenye Mou
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China; (C.M.); (J.X.)
| | - Jiayi Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China; (C.M.); (J.X.)
| | - Wei Cui
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China; (C.M.); (J.X.)
| | - Yijing Shi
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| | - Yangzhe Wang
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| | - Tian Luo
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; (T.L.); (W.G.); (J.Y.)
| | - Wei Guo
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; (T.L.); (W.G.); (J.Y.)
| | - Jichun Ye
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; (T.L.); (W.G.); (J.Y.)
| | - Wanghua Chen
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| |
Collapse
|
36
|
Rout SK, Rhyner D, Greenwald J, Riek R. Characterization of self-templating catalytic amyloids. Methods Enzymol 2024; 697:51-75. [PMID: 38816135 DOI: 10.1016/bs.mie.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Amyloid aggregates with unique periodic structures have garnered significant attention due to their association with numerous diseases, including systemic amyloidoses and the neurodegenerative diseases Parkinson's, Alzheimer's, and Creutzfeld-Jakob. However, more recent investigations have expanded our understanding of amyloids, revealing their diverse functional biological roles. Amyloids have also been proposed to have played a significant role in prebiotic molecular evolution because of their exceptional stability, spontaneous formation in a prebiotic environment, catalytic and templating abilities, and cooperative interaction with fatty acids, polysaccharides, and nucleic acids. This chapter summarizes methods and techniques associated with studying short amyloidogenic peptides, including detailed procedures for investigating cross-templating and autocatalytic templating reactions. Since the work with amyloidogenic peptides and their aggregates present unique challenges, we have attempted to address these with essential details throughout the procedures. The lessons herein may be used in any amyloid-related research to ensure more reproducible results and reduce entrance barriers for researchers new to the field.
Collapse
Affiliation(s)
- Saroj K Rout
- Institute of Molecular Physical Science, ETH Zürich, Zürich, Switzerland; Systems Biophysics, Department of Physics, Ludwig Maximilian University Munich, Munich, Germany
| | - David Rhyner
- Institute of Molecular Physical Science, ETH Zürich, Zürich, Switzerland
| | - Jason Greenwald
- Institute of Molecular Physical Science, ETH Zürich, Zürich, Switzerland.
| | - Roland Riek
- Institute of Molecular Physical Science, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
37
|
Baghel D, de Oliveira AP, Satyarthy S, Chase WE, Banerjee S, Ghosh A. Structural characterization of amyloid aggregates with spatially resolved infrared spectroscopy. Methods Enzymol 2024; 697:113-150. [PMID: 38816120 PMCID: PMC11147165 DOI: 10.1016/bs.mie.2024.02.013] [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] [Indexed: 06/01/2024]
Abstract
The self-assembly of proteins and peptides into ordered structures called amyloid fibrils is a hallmark of numerous diseases, impacting the brain, heart, and other organs. The structure of amyloid aggregates is central to their function and thus has been extensively studied. However, the structural heterogeneities between aggregates as they evolve throughout the aggregation pathway are still not well understood. Conventional biophysical spectroscopic methods are bulk techniques and only report on the average structural parameters. Understanding the structure of individual aggregate species in a heterogeneous ensemble necessitates spatial resolution on the length scale of the aggregates. Recent technological advances have led to augmentation of infrared (IR) spectroscopy with imaging modalities, wherein the photothermal response of the sample upon vibrational excitation is leveraged to provide spatial resolution beyond the diffraction limit. These combined approaches are ideally suited to map out the structural heterogeneity of amyloid ensembles. AFM-IR, which integrates IR spectroscopy with atomic force microscopy enables identification of the structural facets the oligomers and fibrils at individual aggregate level with nanoscale resolution. These capabilities can be extended to chemical mapping in diseased tissue specimens with submicron resolution using optical photothermal microscopy, which combines IR spectroscopy with optical imaging. This book chapter provides the basic premise of these novel techniques and provides the typical methodology for using these approaches for amyloid structure determination. Detailed procedures pertaining to sample preparation and data acquisition and analysis are discussed and the aggregation of the amyloid β peptide is provided as a case study to provide the reader the experimental parameters necessary to use these techniques to complement their research efforts.
Collapse
Affiliation(s)
- Divya Baghel
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL, United States
| | - Ana Pacheco de Oliveira
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL, United States
| | - Saumya Satyarthy
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL, United States
| | - William E Chase
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL, United States
| | - Siddhartha Banerjee
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL, United States
| | - Ayanjeet Ghosh
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL, United States.
| |
Collapse
|
38
|
Lavatelli F, Natalello A, Marchese L, Ami D, Corazza A, Raimondi S, Mimmi MC, Malinverni S, Mangione PP, Palmer MT, Lampis A, Concardi M, Verona G, Canetti D, Arbustini E, Bellotti V, Giorgetti S. Truncation of the constant domain drives amyloid formation by immunoglobulin light chains. J Biol Chem 2024; 300:107174. [PMID: 38499153 PMCID: PMC11016911 DOI: 10.1016/j.jbc.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: 12/08/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
AL amyloidosis is a life-threatening disease caused by deposition of immunoglobulin light chains. While the mechanisms underlying light chains amyloidogenesis in vivo remain unclear, several studies have highlighted the role that tissue environment and structural amyloidogenicity of individual light chains have in the disease pathogenesis. AL natural deposits contain both full-length light chains and fragments encompassing the variable domain (VL) as well as different length segments of the constant region (CL), thus highlighting the relevance that proteolysis may have in the fibrillogenesis pathway. Here, we investigate the role of major truncated species of the disease-associated AL55 light chain that were previously identified in natural deposits. Specifically, we study structure, molecular dynamics, thermal stability, and capacity to form fibrils of a fragment containing both the VL and part of the CL (133-AL55), in comparison with the full-length protein and its variable domain alone, under shear stress and physiological conditions. Whereas the full-length light chain forms exclusively amorphous aggregates, both fragments generate fibrils, although, with different kinetics, aggregate structure, and interplay with the unfragmented protein. More specifically, the VL-CL 133-AL55 fragment entirely converts into amyloid fibrils microscopically and spectroscopically similar to their ex vivo counterpart and increases the amorphous aggregation of full-length AL55. Overall, our data support the idea that light chain structure and proteolysis are both relevant for amyloidogenesis in vivo and provide a novel biocompatible model of light chain fibrillogenesis suitable for future mechanistic studies.
Collapse
Affiliation(s)
- Francesca Lavatelli
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Antonino Natalello
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.
| | - Loredana Marchese
- Pathology Unit, Fondazione IRCSS Policlinico San Matteo, Pavia, Italy
| | - Diletta Ami
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Alessandra Corazza
- Department of Medicine (DAME), University of Udine, Udine, Italy; Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Sara Raimondi
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
| | - Maria Chiara Mimmi
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Silvia Malinverni
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
| | - P Patrizia Mangione
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Manel Terrones Palmer
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Alessio Lampis
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
| | - Monica Concardi
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Guglielmo Verona
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Centre for Amyloidosis, Division of Medicine, University College London, London, UK
| | - Diana Canetti
- Centre for Amyloidosis, Division of Medicine, University College London, London, UK
| | - Eloisa Arbustini
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Vittorio Bellotti
- Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sofia Giorgetti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy; Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
39
|
Nayak K, Sarkar N, Bauri K, De P. Helical Superstructures from the Hierarchical Self-Assembly of Coil-Coil Block Copolymer Guided by Side Chain Amyloid-β(17-19) LVF Peptide. Biomacromolecules 2024; 25:1978-1988. [PMID: 38345926 DOI: 10.1021/acs.biomac.3c01379] [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: 03/12/2024]
Abstract
The rational design of precisely controlled hierarchical chiral nanostructures from synthetic polymers garnered inspiration from sophisticated biological materials. Since chiral peptide motifs induce helix formation in macromolecules, herein we report the synthesis of a novel type of hybrid polymer consisting of a β-sheet forming a LVF [L = leucine, V = valine, and F = phenylalanine] tripeptide pendant polymethacrylate block and a poly[poly(ethylene glycol) methyl ether methacrylate] (PPEGMA) block. The designed block copolymer self-organized into helical superstructures with a left-handed twisting sense, as visualized by field emission scanning electron microscopy, transmission electron microscopy, and atomic force microscopy. This intriguing hierarchical self-assembly is driven by the minimalistic peptide motif that itself has a high propensity to adopt an antiparallel β-sheet conformation. We also report the generation of a diverse array of nanostructures, including spherical micelles, spindle micelles, rod-like micelles, vesicles, helical supramolecular fibers, and helical toroids via self-assembly of the designed block copolymer in tetrahydrofuran/water mixed solvents. To realize the observable helical superstructure, a twisted two-dimensional core-shell tape is proposed as a structure model in which the peptide segments form an antiparallel β-sheet with a polymer shell. The findings contribute to the advancement of a helical polymer or the superhelical self-assembly of polymers, paving the way for diverse applications in materials science and related fields.
Collapse
Affiliation(s)
- Kasturee Nayak
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India
| | - Niharendra Sarkar
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India
| | - Kamal Bauri
- Department of Chemistry, Raghunathpur College, Raghunathpur - 723133, Purulia, West Bengal, India
| | - Priyadarsi De
- Polymer Research Centre and Centre for Advanced Functional Materials, Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur - 741246, Nadia, West Bengal, India
| |
Collapse
|
40
|
Wang Y, Wang Z, Yang L, Zhang W, Ma G. Unravelling the non-classical nucleation mechanism of an amyloid nanosheet through atomic force microscopy and an infrared probe technique. Phys Chem Chem Phys 2024; 26:7855-7864. [PMID: 38376417 DOI: 10.1039/d3cp05345h] [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: 02/21/2024]
Abstract
Understanding the amyloid nucleation mechanism is fundamentally important for the development of diagnostics and therapeutics of amyloid-related diseases and for the design and application of amyloid-based materials. To this end, we here explore the use of atomic force microscopy (AFM) and a side-chain-based infrared (IR) probe technique to investigate the amyloid nanosheet formation mechanism of an Aβ16-22 variant, KLVFXAK, where X is p-cyanophenylalanine with its side-chain cyano group being an infrared probe. Using AFM, we reveal that the formation of KLVFXAK amyloid nanosheets follows a two-step non-classical nucleation mechanism. The first step is the rapid formation of a metastable fibrillar intermediate and the second step is slow transformation to the final nanosheet. Using the side-chain-based IR probe technique, we obtain spectroscopic evidence for the proposed nucleation mechanism of the amyloid nanosheet as well as the structural details for the intermediate and amyloid nanosheet. By using the structural constraints set by the two techniques, we propose the structural models for both the fibrillar intermediate and the amyloid nanosheet. In addition, we further investigated the amyloid nanosheet formation mechanism of a similar Aβ16-22 variant, KLVFXAE, and showed the impact of mutation on the amyloid nucleation mechanism. Our work also provides a nice example of how to use the combined approach of AFM and a side-chain-based IR probe technique to unravel the complex nucleation mechanism of amyloid formation.
Collapse
Affiliation(s)
- Yao Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China.
| | - Ziqi Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China.
| | - Lujuan Yang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China.
| | - Wenkai Zhang
- Department of Physics, Applied Optics Beijing Area Major Laboratory, Center for Advanced Quantum Studies, Beijing Normal University, Beijing 100875, China.
| | - Gang Ma
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China.
| |
Collapse
|
41
|
Suresh K, Dahal E, Badano A. Synthetic β-sheets mimicking fibrillar and oligomeric structures for evaluation of spectral X-ray scattering technique for biomarker quantification. Cell Biosci 2024; 14:26. [PMID: 38374092 PMCID: PMC10877803 DOI: 10.1186/s13578-024-01208-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Archetypical cross-β spines sharpen the boundary between functional and pathological proteins including β-amyloid, tau, α-synuclein and transthyretin are linked to many debilitating human neurodegenerative and non-neurodegenerative amyloidoses. An increased focus on development of pathogenic β-sheet specific fluid and imaging structural biomarkers and conformation-specific monoclonal antibodies in targeted therapies has been recently observed. Identification and quantification of pathogenic oligomers remain challenging for existing neuroimaging modalities. RESULTS We propose two artificial β-sheets which can mimic the nanoscopic structural characteristics of pathogenic oligomers and fibrils for evaluating the performance of a label free, X-ray based biomarker detection and quantification technique. Highly similar structure with elliptical cross-section and parallel cross-β motif is observed among recombinant α-synuclein fibril, Aβ-42 fibril and artificial β-sheet fibrils. We then use these β-sheet models to assess the performance of spectral small angle X-ray scattering (sSAXS) technique for detecting β-sheet structures. sSAXS showed quantitatively accurate detection of antiparallel, cross-β artificial oligomers from a tissue mimicking environment and significant distinction between different oligomer packing densities such as diffuse and dense packings. CONCLUSION The proposed synthetic β-sheet models mimicked the nanoscopic structural characteristics of β-sheets of fibrillar and oligomeric states of Aβ and α-synuclein based on the ATR-FTIR and SAXS data. The tunability of β-sheet proportions and shapes of structural motifs, and the low-cost of these β-sheet models can become useful test materials for evaluating β-sheet or amyloid specific biomarkers in a wide range of neurological diseases. By using the proposed synthetic β-sheet models, our study indicates that the sSAXS has potential to evaluate different stages of β-sheet-enriched structures including oligomers of pathogenic proteins.
Collapse
Affiliation(s)
- Karthika Suresh
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD, 20993, USA.
| | - Eshan Dahal
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Aldo Badano
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD, 20993, USA
| |
Collapse
|
42
|
Kino S, Kanamori M, Shimoda Y, Niizuma K, Endo H, Matsuura Y. Distinguishing IDH mutation status in gliomas using FTIR-ATR spectra of peripheral blood plasma indicating clear traces of protein amyloid aggregation. BMC Cancer 2024; 24:222. [PMID: 38365669 PMCID: PMC10870484 DOI: 10.1186/s12885-024-11970-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Glioma is a primary brain tumor and the assessment of its molecular profile in a minimally invasive manner is important in determining treatment strategies. Among the molecular abnormalities of gliomas, mutations in the isocitrate dehydrogenase (IDH) gene are strong predictors of treatment sensitivity and prognosis. In this study, we attempted to non-invasively diagnose glioma development and the presence of IDH mutations using multivariate analysis of the plasma mid-infrared absorption spectra for a comprehensive and sensitive view of changes in blood components associated with the disease and genetic mutations. These component changes are discussed in terms of absorption wavenumbers that contribute to differentiation. METHODS Plasma samples were collected at our institutes from 84 patients with glioma (13 oligodendrogliomas, 17 IDH-mutant astrocytoma, 7 IDH wild-type diffuse glioma, and 47 glioblastomas) before treatment initiation and 72 healthy participants. FTIR-ATR spectra were obtained for each plasma sample, and PLS discriminant analysis was performed using the absorbance of each wavenumber in the fingerprint region of biomolecules as the explanatory variable. This data was used to distinguish patients with glioma from healthy participants and diagnose the presence of IDH mutations. RESULTS The derived classification algorithm distinguished the patients with glioma from healthy participants with 83% accuracy (area under the curve (AUC) in receiver operating characteristic (ROC) = 0.908) and diagnosed the presence of IDH mutation with 75% accuracy (AUC = 0.752 in ROC) in cross-validation using 30% of the total test data. The characteristic changes in the absorption spectra suggest an increase in the ratio of β-sheet structures in the conformational composition of blood proteins of patients with glioma. Furthermore, these changes were more pronounced in patients with IDH-mutant gliomas. CONCLUSIONS The plasma infrared absorption spectra could be used to diagnose gliomas and the presence of IDH mutations in gliomas with a high degree of accuracy. The spectral shape of the protein absorption band showed that the ratio of β-sheet structures in blood proteins was significantly higher in patients with glioma than in healthy participants, and protein aggregation was a distinct feature in patients with glioma with IDH mutations.
Collapse
Affiliation(s)
- Saiko Kino
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-05, Aza-Aoba, Aramaki, Aoba, Sendai City, 980-8579, Miyagi Prefecture, Japan
| | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, 980-8574 Seiryo 1-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Yoshiteru Shimoda
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, 980-8574 Seiryo 1-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Seiryo 2-1, Aoba, Sendai City, 980-8575, Miyagi Prefecture, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, 980-8575 Seiryo 2-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, 980-8574 Seiryo 1-1, Aoba, Sendai City, Miyagi Prefecture, Japan
| | - Yuji Matsuura
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-05, Aza-Aoba, Aramaki, Aoba, Sendai City, 980-8579, Miyagi Prefecture, Japan.
| |
Collapse
|
43
|
Tsai TY, Jhang WT, Hsu HK, Chan YT, Chang CF, Chen YR. Amyloid Modifier SERF1a Accelerates Alzheimer's Amyloid-β Fibrillization and Exacerbates the Cytotoxicity. ACS Chem Neurosci 2024; 15:479-490. [PMID: 38211979 DOI: 10.1021/acschemneuro.3c00403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating, progressive neurodegenerative disease affecting the elderly in the world. The pathological hallmark senile plaques are mainly composed of amyloid-β (Aβ), in which the main isoforms are Aβ40 and Aβ42. Aβ is prone to aggregate and ultimately forms amyloid fibrils in the brains of AD patients. Factors that alter the Aβ aggregation process have been considered to be potential targets for treatments of AD. Modifier of aggregation 4 (MOAG-4)/small EDRK-rich factor (SERF) was previously selected from a chemical mutagenesis screen and identified as an amyloid modifier that promotes amyloid aggregation for α-synuclein, huntingtin, and Aβ40. The interaction and effect of yeast ScSERF on Aβ40 were previously described. Here, we examined the human SERF1a effect on Aβ40 and Aβ42 fibrillization by the Thioflavin T assay and found that SERF1a accelerated Aβ fibrillization in a dose-dependent manner without changing the fibril amount and without incorporation. By Fourier transform infrared spectroscopy (FTIR) and transmission electron microscopy (TEM), we found that SERF1a altered the secondary structures and the morphology of Aβ fibrils. The electrospray ionization mass spectrometry (ESI-MS) and analytical ultracentrifugation (AUC) results showed that SERF1a binds to Aβ in a 1:1 stoichiometry. Moreover, the NMR study showed that SERF1a interacts with Aβ via its N-terminal region. Cytotoxicity assay demonstrated that SERF1a enhanced toxicity of Aβ intermediates, and the effect can be rescued by SERF1a antibody. Overall, our study provides the underlying molecular mechanism for the SERF1a effect on Aβ fibrillization and facilitates the therapeutic development of AD.
Collapse
Affiliation(s)
- Tien-Ying Tsai
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 115, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Biological Chemistry, Academia Sinica, 128, Academia Road, Sec. 2, Nankang District, Taipei 115, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Wei-Ting Jhang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Hung-Kai Hsu
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yi-Tsu Chan
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Chi-Fon Chang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 115, Taiwan
| | - Yun-Ru Chen
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 115, Taiwan
| |
Collapse
|
44
|
Siri M, Mangiarotti A, Vázquez-Dávila M, Bidan CM. Curli Amyloid Fibers in Escherichia coli Biofilms: The Influence of Water Availability on their Structure and Functional Properties. Macromol Biosci 2024; 24:e2300234. [PMID: 37776075 DOI: 10.1002/mabi.202300234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/22/2023] [Indexed: 10/01/2023]
Abstract
Escherichia coli biofilms consist of bacteria embedded in a self-produced matrix mainly made of protein fibers and polysaccharides. The curli amyloid fibers found in the biofilm matrix are promising versatile building blocks to design sustainable bio-sourced materials. To exploit this potential, it is crucial to understand i) how environmental cues during biofilm growth influence the molecular structure of these amyloid fibers, and ii) how this translates at higher length scales. To explore these questions, the effect of water availability during biofilm growth on the conformation and functions of curli is studied. Microscopy and spectroscopy are used to characterize the amyloid fibers purified from biofilms grown on nutritive substrates with different water contents, and micro-indentation to measure the rigidity of the respective biofilms. The purified curli amyloid fibers present differences in the yield, structure, and functional properties upon biofilm growth conditions. Fiber packing and β-sheets content correlate with their hydrophobicity and chemical stability, and with the rigidity of the biofilms. This study highlights how E. coli biofilm growth conditions impact curli structure and functions contributing to macroscopic materials properties. These fundamental findings infer an alternative strategy to tune curli structure, which will ultimately benefit engineering hierarchical and functional curli-based materials.
Collapse
Affiliation(s)
- Macarena Siri
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, 14476, Potsdam, Germany
| | - Agustín Mangiarotti
- Max Planck Institute of Colloids and Interfaces, Department of Sustainable and Bio-inspired Materials, 14476, Potsdam, Germany
| | - Mónica Vázquez-Dávila
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, 14476, Potsdam, Germany
| | - Cécile M Bidan
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, 14476, Potsdam, Germany
| |
Collapse
|
45
|
de Oliveira AP, Chase W, Confer MP, Walker S, Baghel D, Ghosh A. Colocalization of β-Sheets and Carotenoids in Aβ Plaques Revealed with Multimodal Spatially Resolved Vibrational Spectroscopy. J Phys Chem B 2024; 128:33-44. [PMID: 38124262 PMCID: PMC10851346 DOI: 10.1021/acs.jpcb.3c04782] [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] [Indexed: 12/23/2023]
Abstract
The aggregation of amyloid β(Aβ) peptides is at the heart of Alzheimer's disease development and progression. As a result, amyloid aggregates have been studied extensively in vitro, and detailed structural information on fibrillar amyloid aggregates is available. However, forwarding these structural models to amyloid plaques in the human brain is still a major challenge. The chemistry of amyloid plaques, particularly in terms of the protein secondary structure and associated chemical moieties, remains poorly understood. In this report, we use Raman microspectroscopy to identify the presence of carotenoids in amyloid plaques and demonstrate that the abundance of carotenoids is correlated with the overall protein secondary structure of plaques, specifically to the population of β-sheets. While the association of carotenoids with plaques has been previously identified, their correlation with the β structure has never been identified. To further validate these findings, we have used optical photothermal infrared (O-PTIR) spectroscopy, which is a spatially resolved technique that yields complementary infrared contrast to Raman. O-PTIR unequivocally demonstrates the presence of elevated β-sheets in carotenoid-containing plaques and the lack of β structure in noncarotenoid plaques. Our findings underscore the potential link between anti-inflammatory species as carotenoids to specific secondary structural motifs within Aβ plaques and highlight the possible role of chemically distinct plaques in neuroinflammation, which can uncover new mechanistic insights and lead to new therapeutic strategies for AD.
Collapse
Affiliation(s)
| | - William Chase
- Department of Chemistry and Biochemistry, University of Alabama, Tuscaloosa, AL 35401, USA
| | - Matthew P. Confer
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana Champaign, Urbana, Illinois 61801, USA
| | - Savannah Walker
- Department of Chemistry and Biochemistry, University of Alabama, Tuscaloosa, AL 35401, USA
| | - Divya Baghel
- Department of Chemistry and Biochemistry, University of Alabama, Tuscaloosa, AL 35401, USA
| | - Ayanjeet Ghosh
- Department of Chemistry and Biochemistry, University of Alabama, Tuscaloosa, AL 35401, USA
| |
Collapse
|
46
|
Yang L, Wang Y, Zhang W, Ma G. New Insight into the Structural Nature of Diphenylalanine Nanotube through Comparison with Amyloid Assemblies. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:1046-1057. [PMID: 38153333 DOI: 10.1021/acs.langmuir.3c03270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Diphenylalanine (FF) nanotubes are a star material in the field of peptide self-assembly and have demonstrated numerous intriguing applications. Due to its resemblance to amyloid assembly, the FF nanotube is widely regarded as a simplified mimic of amyloids. Yet, whether FF nanotube truly possesses amyloid structure remains an open question. To better understand the structural nature of FF nanotube, we herein performed a comparative structural investigation between FF nanotube and typical amyloid systems by Aβ1-40, Aβ1-42, Aβ16-22, Aβ13-23, α-synuclein, and lysozyme using Fourier transform infrared spectroscopy. Through this comparative investigation, we obtained clear evidence to support that the FF nanotube does not possess a β-sheet structure, a key structural characteristic of amyloid assembly, thus revealing the non-amyloid structural nature of the FF nanotube. At last, in light of our new finding, we further discussed the unique self-assembly behaviors of FF during nanotube formation and the implications of our work for FF nanotube related applications.
Collapse
Affiliation(s)
- Lujuan Yang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China
| | - Yao Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China
| | - Wenkai Zhang
- Department of Physics, Applied Optics Beijing Area Major Laboratory, Center for Advanced Quantum Studies, Beijing Normal University, Beijing 100875, China
| | - Gang Ma
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China
| |
Collapse
|
47
|
Lucignano R, Spadaccini R, Merlino A, Ami D, Natalello A, Ferraro G, Picone D. Structural insights and aggregation propensity of a super-stable monellin mutant: A new potential building block for protein-based nanostructured materials. Int J Biol Macromol 2024; 254:127775. [PMID: 38287601 DOI: 10.1016/j.ijbiomac.2023.127775] [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: 08/28/2023] [Revised: 10/12/2023] [Accepted: 10/27/2023] [Indexed: 01/31/2024]
Abstract
Protein fibrillation is commonly associated with pathologic amyloidosis. However, under appropriate conditions several proteins form fibrillar structures in vitro that can be used for biotechnological applications. MNEI and its variants, firstly designed as single chain derivatives of the sweet protein monellin, are also useful models for protein fibrillary aggregation studies. In this work, we have drawn attention to a protein dubbed Mut9, already characterized as a "super stable" MNEI variant. Comparative analysis of the respective X-ray structures revealed how the substitutions present in Mut9 eliminate several unfavorable interactions and stabilize the global structure. Molecular dynamic predictions confirmed the presence of a hydrogen-bonds network in Mut9 which increases its stability, especially at neutral pH. Thioflavin-T (ThT) binding assays and Fourier transform infrared (FTIR) spectroscopy indicated that the aggregation process occurs both at acidic and neutral pH, with and without addition of NaCl, even if with a different kinetics. Accordingly, Transmission Electron Microscopy (TEM) showed a fibrillar organization of the aggregates in all the tested conditions, albeit with some differences in the quantity and in the morphology of the fibrils. Our data underline the great potential of Mut9, which combines great stability in solution with the versatile conversion into nanostructured biomaterials.
Collapse
Affiliation(s)
- Rosanna Lucignano
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia, 80126 Naples, Italy
| | - Roberta Spadaccini
- Department of Science and Technology, University of Sannio, Via de Sanctis, 82100 Benevento, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia, 80126 Naples, Italy
| | - Diletta Ami
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 20126 Milano, Italy
| | - Antonino Natalello
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 20126 Milano, Italy
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia, 80126 Naples, Italy.
| | - Delia Picone
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia, 80126 Naples, Italy.
| |
Collapse
|
48
|
Prabhu MPT, Chrungoo S, Sarkar N. Carboxylic Group Functionalized Carbon Quantum Dots inhibit Hen Egg White Lysozyme Amyloidogenesis, leading to the Formation of Spherical Aggregates with Reduced Toxicity and ROS Generation. Curr Protein Pept Sci 2024; 25:626-637. [PMID: 38659260 DOI: 10.2174/0113892037294778240328041907] [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: 11/26/2023] [Revised: 02/12/2024] [Accepted: 02/29/2024] [Indexed: 04/26/2024]
Abstract
INTRODUCTION Proteinopathies are a group of diseases where the protein structure has been altered. These alterations are linked to the production of amyloids, which are persistent, organized clumps of protein molecules through inter-molecular interactions. Several disorders, including Alzheimer's and Parkinson's, have been related to the presence of amyloids. Highly ordered beta sheets or beta folds are characteristic of amyloids; these structures can further self- assemble into stable fibrils. METHODS Protein aggregation is caused by a wide variety of environmental and experimental factors, including mutations, high pH, high temperature, and chemical modification. Despite several efforts, a cure for amyloidosis has yet to be found. Due to its advantageous semi-conducting characteristics, unique optical features, high surface area-to-volume ratio, biocompatibility, etc., carbon quantum dots (CQDs) have lately emerged as key instruments for a wide range of biomedical applications. To this end, we have investigated the effect of CQDs with a carboxyl group on their surface (CQD-CA) on the in vitro amyloidogenesis of hen egg white lysozyme (HEWL). RESULTS By generating a stable compound that is resistant to fibrillation, our findings show that CQD-CA can suppress amyloid and disaggregate HEWL. In addition, CQD-CA caused the creation of non-toxic spherical aggregates, which generated much less reactive oxygen species (ROS). CONCLUSION Overall, our results show that more research into amyloidosis treatments, including surface functionalized CQDs, is warranted.
Collapse
Affiliation(s)
- M P Taraka Prabhu
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Shreya Chrungoo
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Nandini Sarkar
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| |
Collapse
|
49
|
Siraj S, Yameen D, Bhati S, Athar T, Khan S, Bhattacharya J, Islam A, Haque MM. Sugar osmolyte inhibits and attenuates the fibrillogenesis in RNase A: An in vitro and in silico characterizations. Int J Biol Macromol 2023; 253:127378. [PMID: 37839601 DOI: 10.1016/j.ijbiomac.2023.127378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/07/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Mechanisms of protein aggregation are of immense interest in therapeutic biology and neurodegenerative medicine. Biochemical processes within the living cell occur in a highly crowded environment. The phenomenon of macromolecular crowding affects the diffusional and conformational dynamics of proteins and modulates their folding. Macromolecular crowding is reported to cause protein aggregation in some cases, so it is a cause of concern as it leads to a plethora of neurodegenerative disorders and systemic amyloidosis. To divulge the mechanism of aggregation, it is imperative to study aggregation in well-characterized model proteins in the presence of macromolecular crowder. One such protein is ribonuclease A (RNase A), which deciphers neurotoxic function in humans; therefore we decided to explore the amyloid fibrillogenesis of this thermodynamically stable protein. To elucidate the impact of crowder, dextran-70 and its monomer glucose on the aggregation profile of RNase-A various techniques such as Absorbance, Fluorescence, Fourier Transforms Infrared, Dynamic Light Scattering and circular Dichroism spectroscopies along with imaging techniques like Atomic Force Microscopy and Transmission Electron Microscopy were employed. Thermal aggregation and fibrillation were further promoted by dextran-70 while glucose counteracted the effect of the crowding agent in a concentration-dependent manner. This study shows that glucose provides stability to the protein and prevents fibrillation. Intending to combat aggregation, which is the hallmark of numerous late-onset neurological disorders and systemic amyloidosis, this investigation unveils that naturally occurring osmolytes or other co-solutes can be further exploited in novel drug design strategies.
Collapse
Affiliation(s)
- Seerat Siraj
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Daraksha Yameen
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Shivani Bhati
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Teeba Athar
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Salman Khan
- Translational Research Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | | | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Mohammad Mahfuzul Haque
- Molecular Enzymology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
50
|
Smeralda W, Since M, Corvaisier S, Fayolle D, Cardin J, Duprey S, Jourdan JP, Cullin C, Malzert-Freon A. A Biomimetic Multiparametric Assay to Characterise Anti-Amyloid Drugs. Int J Mol Sci 2023; 24:16982. [PMID: 38069305 PMCID: PMC10707238 DOI: 10.3390/ijms242316982] [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: 10/27/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most widespread form of senile dementia worldwide and represents a leading socioeconomic problem in healthcare. Although it is widely debated, the aggregation of the amyloid β peptide (Aβ) is linked to the onset and progression of this neurodegenerative disease. Molecules capable of interfering with specific steps in the fibrillation process remain of pharmacological interest. To identify such compounds, we have set up a small molecule screening process combining multiple experimental methods (UV and florescence spectrometry, ITC, and ATR-FTIR) to identify and characterise potential modulators of Aβ1-42 fibrillation through the description of the biochemical interactions (molecule-membrane Aβ peptide). Three known modulators, namely bexarotene, Chicago sky blue and indomethacin, have been evaluated through this process, and their modulation mechanism in the presence of a biomembrane has been described. Such a well-adapted physico-chemical approach to drug discovery proves to be an undeniable asset for the rapid characterisation of compounds of therapeutic interest for Alzheimer's disease. This strategy could be adapted and transposed to search for modulators of other amyloids such as tau protein.
Collapse
Affiliation(s)
- Willy Smeralda
- Normandie Université, UNICAEN, CERMN, Boulevard Becquerel, 14000 Caen, France; (W.S.); (S.C.); (D.F.); (J.-P.J.)
| | - Marc Since
- Normandie Université, UNICAEN, CERMN, Boulevard Becquerel, 14000 Caen, France; (W.S.); (S.C.); (D.F.); (J.-P.J.)
| | - Sophie Corvaisier
- Normandie Université, UNICAEN, CERMN, Boulevard Becquerel, 14000 Caen, France; (W.S.); (S.C.); (D.F.); (J.-P.J.)
| | - Dimitri Fayolle
- Normandie Université, UNICAEN, CERMN, Boulevard Becquerel, 14000 Caen, France; (W.S.); (S.C.); (D.F.); (J.-P.J.)
| | - Julien Cardin
- CIMAP, ENSICAEN, UNICAEN, UMR6252 CNRS, CEA, Normandie Université, 6 Bd du Maréchal Juin, 14050 Caen, France; (J.C.); (S.D.)
| | - Sylvain Duprey
- CIMAP, ENSICAEN, UNICAEN, UMR6252 CNRS, CEA, Normandie Université, 6 Bd du Maréchal Juin, 14050 Caen, France; (J.C.); (S.D.)
| | - Jean-Pierre Jourdan
- Normandie Université, UNICAEN, CERMN, Boulevard Becquerel, 14000 Caen, France; (W.S.); (S.C.); (D.F.); (J.-P.J.)
- Pharmacie à Usage Intérieur, Centre Hospitalier de Vire, Normandie, 14504 Vire, France
| | | | - Aurélie Malzert-Freon
- Normandie Université, UNICAEN, CERMN, Boulevard Becquerel, 14000 Caen, France; (W.S.); (S.C.); (D.F.); (J.-P.J.)
| |
Collapse
|