1
|
Dolui S, Maity A, Kundu S, Nanda B, Roy A, Mondal A, Adhikary A, Saha A, Pal U, Bhunia A, Maiti NC. Stabilization of α-Helical Folded Structures Retards Hydrophobic Zipping and Fibrillation of Bovine Insulin: A Key Signature from Raman Spectroscopic Analysis. J Phys Chem B 2025. [PMID: 40289529 DOI: 10.1021/acs.jpcb.5c00846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Insulin is an α-helical-rich globular protein that is well-stabilized via several noncovalent forces including the inter-residue/intersubunit hydrophobic interactions. However, similar noncovalent forces, although of different degrees and orientations, effectuate many proteins to assemble and adapt thermodynamically stable β-sheet-rich fibrillar aggregates, causing a severe impact on their native structure and function. This fibrillation of proteins involves a key event, which is the zipping of hydrophobic amyloidogenic regions that are exposed intrinsically or become bared in the folded proteins under harsh conditions. This study has revealed that Coomassie Brilliant Blue G-250 (CBBG) can inhibit the essential zipping processes and stabilize the α-helical structure of bovine insulin (BI), resulting in a significant delay in the fibril formation. The interaction of CBBG with BI was found to be a thermodynamically favorable event, with it being an enthalpy-driven process (ΔH0 -88.04 kcal/mol), with the change in Gibb's free energy (ΔG0) observed to be ∼ -6.98 kcal/mol. Surface-enhanced Raman scattering measurements showed a characteristic α-helical signal of the protein at 1649 cm-1 in the presence of CBBG, suggesting the enhanced thermal stability of the hormone. Computational analysis further revealed that CBBG binds to both chains A and B of bovine insulin and boosts the folding stability in the monomeric state, causing a significant reduction in its structural fluctuation. The sulfonate moieties of CBBG showed significant intermolecular interactions with the B chain of N-terminal segments. Specifically, one sulfonate group formed multiple hydrogen bonds with both the backbone amide group and the terminal amine. Also, the N-terminal phenylalanine residue of BI (F1B) was found to have a significant contribution to the hydrophobic π-π stacking interactions with the CBBG aromatic phenyl ring.
Collapse
Affiliation(s)
- Sandip Dolui
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, 4, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Anupam Maity
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, 4, Raja S. C. Mallick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Shubham Kundu
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, 4, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Banadipa Nanda
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, 4, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Anupam Roy
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, 4, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Animesh Mondal
- Zoology, Govt. Gen. De. College, Mangalkote, Panchanantala, Khudrun, Purba Bardhaman, West Bengal 713132, India
| | - Ananya Adhikary
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, 4, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, 92, Acharya Prafulla Chandra Road, Calcutta 700009, India
| | - Uttam Pal
- S. N. Bose National Centre for Basic Sciences, Technical Research Centre, Kolkata 700106, India
| | - Anirban Bhunia
- Department of Chemical Sciences, Bose Institute, Unified Academic Campus, Salt Lake, EN80, Kolkata 700091, India
| | - Nakul C Maiti
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, 4, Raja S. C. Mallick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| |
Collapse
|
2
|
Hima S, Aiswarya N, Remya C, Vasudevan DM, Dileep KV, Francis D. Deciphering protein aggregation: Insights into morphology, contributing factors, and molecular pathologies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 145:23-71. [PMID: 40324848 DOI: 10.1016/bs.apcsb.2024.11.009] [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: 05/07/2025]
Abstract
Protein aggregation research stands at the cutting edge of biomedical science, offering crucial insights into the molecular underpinnings of neurodegenerative and amyloid-associated diseases. Significant advancements in deciphering the structural, biophysical, and molecular intricacies of protein misfolding are driving the development of innovative therapies. Emerging approaches, from small molecule inhibitors to sophisticated polymer-based therapeutics, hold great promise for alleviating the toxic impacts of aggregation with the potential to prevent, delay, or even reverse disease progression. Despite these advances, the field contends with substantial challenges. The polymorphic and complex nature of protein aggregates poses major obstacles to both research and therapeutic design. Yet, interdisciplinary methodologies-integrating advanced spectroscopic, imaging, and computational tools-are creating new pathways to address these complexities, effectively bridging molecular breakthroughs and practical therapeutic applications. The rapid shift of foundational discoveries to clinical trials marks a pivotal step forward, instilling new hope for patients with protein aggregation disorders. Each breakthrough propels us closer to life-changing therapies that may reshape the outlook for these patients. The promise of precise and effective treatments is driving a transformative shift in medical science, establishing protein aggregation research as a crucial pillar in combating these challenging diseases and offering a beacon of hope for the future of neurodegenerative care.
Collapse
Affiliation(s)
- Sree Hima
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India; Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi, India
| | - N Aiswarya
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India; Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Chandran Remya
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India
| | - D M Vasudevan
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India; Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi, India
| | - K V Dileep
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India; Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi, India.
| | - Dileep Francis
- Department of Life sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| |
Collapse
|
3
|
Xia M, Ding L, Ahn DU, Xu L, Huang X, Shu D, Hu W, Cai Z. Whole process of Fab antibody aggregation in intestinal environment and their aggregation regulation: An insight from static and concentration perturbation aggregations. Int J Biol Macromol 2025; 304:140756. [PMID: 39922361 DOI: 10.1016/j.ijbiomac.2025.140756] [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: 08/17/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
This work revealed the aggregation and aggregation inhibition mechanisms of Fab antibody in a simulated intestinal fluid via static and concentration perturbation aggregations to meet the challenges of oral antibody therapy. Results showed that Fab aggregation was highly concentration dependent, mainly determined by the β-sheet's stacking and amyloid fibers' extension at low (1 mg/mL) and the twine of β-strands' turn at high (20 mg/mL) concentrations. During the incubation of 0-240 min, Fab was continuously aggregating, but with some rearrangements on its spatial conformation: α-helix and β-sheet formation with β-turn and random coil unfolding, which expanded the aggregates' hydrophobic core and extended β-sheet structure through the π-π stacking of aromatic amino acids. The aggregation kinetics indicated that high Fab concentrations promoted high aggregates but the growth of amyloid fibers took a long time, while low to high concentration fluctuation promoted the formation of Fab aggregates. Molecular docking and molecular dynamics simulations suggested that Arg, PEG 10000, and Poloxamer 188 reduced the potential energy; PEG 10000 and Tween 20 enhanced steric hindrance by spontaneously binding through competitive hydrogen bonding without disturbing Fab's conformation. This work can provide promising approaches for our daily health management by facilitating the materialization of Fab-based oral antibody therapy.
Collapse
Affiliation(s)
- Minquan Xia
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China
| | - Lixian Ding
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dong Uk Ahn
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Ligen Xu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xi Huang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dewei Shu
- Zaozhuang Key Laboratory of Egg Nutrition and Health, Zaozhuang Jensur Bio-pharmaceutical Co., Ltd, Shandong 277000, China
| | - Wei Hu
- Wuhan Milai Biotechnology Co.,Ltd., Wuhan, Hubei 430000, China
| | - Zhaoxia Cai
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
4
|
More SR, Jha SK. Multi-Site Red-Edge Excitation Shift Reveals the Residue-Specific Solvation Dynamics during the Native to Amyloid-like Transition of an Amyloidogenic Protein. J Phys Chem B 2025; 129:176-193. [PMID: 39682034 DOI: 10.1021/acs.jpcb.4c07067] [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: 12/18/2024]
Abstract
Changes in water-protein interactions are crucial for proteins to achieve functional and nonfunctional conformations during structural transitions by modulating local stability. Amyloid-like protein aggregates in deteriorating neurons are hallmarks of neurodegenerative disorders. These aggregates form through significant structural changes, transitioning from functional native conformations to supramolecular cross-β-sheet structures via misfolded and oligomeric intermediates in a multistep process. However, the site-specific dynamics of water molecules from the native to misfolded conformations and further to oligomeric and compact amyloid structures remain poorly understood. In this study, we used the fluorescence method known as red-edge excitation shift (REES) to investigate the solvation dynamics at specific sites in various equilibrium conformations en route to the misfolding and aggregation of the functional domain of the TDP-43 protein (TDP-43tRRM). We generated three single tryptophan-single cysteine mutants of TDP-43tRRM, with the cysteines at different positions and tryptophan at a fixed position. Each sole cysteine was fluorescently labeled and used as a site-specific fluorophore along with the single tryptophan, creating four monitorable sites for REES studies. By investigating the site-specific extent of REES, we developed a residue-specific solvation dynamics map of TDP-43tRRM during its misfolding and aggregation. Our observations revealed that solvation dynamics progressively became more rigid and heterogeneous to varying extents at different sites during the transition from native to amyloid-like conformations.
Collapse
Affiliation(s)
- Sonal R More
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Kumar Jha
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
5
|
Roe EF, Freire Haddad H, Lazar KM, Liu P, Collier JH. Tuning Helical Peptide Nanofibers as a Sublingual Vaccine Platform for a Variety of Peptide Epitopes. Adv Healthc Mater 2025; 14:e2402055. [PMID: 39676346 PMCID: PMC11949279 DOI: 10.1002/adhm.202402055] [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: 06/04/2024] [Revised: 11/11/2024] [Indexed: 12/17/2024]
Abstract
Mucosal immune responses to vaccination are essential for achieving full protection against pathogens entering their host at mucosal sites. However, traditional parenteral immunization routes commonly fail to raise significant mucosal immunity. Sublingual immunization is a promising alternative delivery route to raise robust immune responses both systemically and at mucosal sites, and nanomaterial-based subunit vaccine platforms offer opportunities for raising epitope-specific responses. Here, sublingual immunization is reported using the Coil29 platform of coiled-coil self-assembling peptide nanofibers. The successful immunization with epitopes of varying physicochemical properties by including mucus-modulating components - namely sequences of proline, alanine, and serine (PAS) is demonstrated. PASylation is shown to decrease mucin complexation and increase epithelial penetration in vitro, enabling sublingual immunization against a variety of selected peptide epitopes in vivo. Coil29 fibers are also readily formed into tablets for solid-state dosing formulations and maintain their immunogenicity in this state. Previous sublingual peptide nanofiber immunotherapies have been based on different structures, such as highly stable β-sheets. The present work demonstrates that alternatively folded structures such as α-helical nanofibers can also be rendered sublingually immunogenic, enabling immunization with a variety of peptide epitopes and offering additional ways to specify mucus interactions, delivery state, dosing, and formulation.
Collapse
Affiliation(s)
- Emily F Roe
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | | | - Kat M Lazar
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Peiying Liu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
6
|
Fakhranurova L, Marchenkov V, Glukhov A, Balobanov V, Ryabova N, Ilyina N, Katina N. Acceleration of carbonic anhydrase amyloid aggregation leads to a decrease in the fibrils toxicity. Biochem Biophys Res Commun 2024; 741:151082. [PMID: 39622157 DOI: 10.1016/j.bbrc.2024.151082] [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: 11/01/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/11/2024]
Abstract
Cells damage by protein aggregates is one of the causes of amyloid diseases. This study aimed to explore the structural features of cytotoxic amyloid fibrils and to find strategies to reduce their damaging effect. Bovine carbonic anhydrase B (BCAB) was chosen for this work due to high toxicity of its amyloid fibrils. The kinetics of amyloid formation, structural features and cytotoxicity of mature fibrils and early globular aggregates formed by wild type protein and six mutant variants have been investigated. The results showed that an increase in residue hydrophobicity accelerates amyloid aggregation, but the formed fibrils have a reduced content of cross-β-structure and are non-toxic to cells. On the contrary, a decrease in hydrophobicity due to the L139A substitution and slowing the initiation of aggregation leads to the formation of highly toxic oligomers during the lag-period. Thus, the data obtained conclude that accelerating amyloid formation can alter the structure of amyloid aggregates and decrease their cytotoxicity.
Collapse
Affiliation(s)
- Liliia Fakhranurova
- Branch of the Institute of Bioorganic Chemistry RAS, Prospekt Nauki, 6, Pushchino, 142290, Russia.
| | - Victor Marchenkov
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Anatoly Glukhov
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Vitalii Balobanov
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Natalya Ryabova
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Nelly Ilyina
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Natalya Katina
- Branch of the Institute of Bioorganic Chemistry RAS, Prospekt Nauki, 6, Pushchino, 142290, Russia; Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| |
Collapse
|
7
|
Choi SI, Jin Y, Choi Y, Seong BL. Beyond Misfolding: A New Paradigm for the Relationship Between Protein Folding and Aggregation. Int J Mol Sci 2024; 26:53. [PMID: 39795912 PMCID: PMC11720324 DOI: 10.3390/ijms26010053] [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: 11/29/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Aggregation is intricately linked to protein folding, necessitating a precise understanding of their relationship. Traditionally, aggregation has been viewed primarily as a sequential consequence of protein folding and misfolding. However, this conventional paradigm is inherently incomplete and can be deeply misleading. Remarkably, it fails to adequately explain how intrinsic and extrinsic factors, such as charges and cellular macromolecules, prevent intermolecular aggregation independently of intramolecular protein folding and structure. The pervasive inconsistencies between protein folding and aggregation call for a new framework. In all combined reactions of molecules, both intramolecular and intermolecular rate (or equilibrium) constants are mutually independent; accordingly, intrinsic and extrinsic factors independently affect both rate constants. This universal principle, when applied to protein folding and aggregation, indicates that they should be treated as two independent yet interconnected processes. Based on this principle, a new framework provides groundbreaking insights into misfolding, Anfinsen's thermodynamic hypothesis, molecular chaperones, intrinsic chaperone-like activities of cellular macromolecules, intermolecular repulsive force-driven aggregation inhibition, proteome solubility maintenance, and proteinopathies. Consequently, this paradigm shift not only refines our current understanding but also offers a more comprehensive view of how aggregation is coupled to protein folding in the complex cellular milieu.
Collapse
Affiliation(s)
- Seong Il Choi
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
| | - Yoontae Jin
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yura Choi
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
- Department of Integrative Biotechnology, Yonsei University, Incheon 21983, Republic of Korea
| | - Baik L. Seong
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
- Department of Microbiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
8
|
Ngah WZW, Ahmad HF, Ankasha SJ, Makpol S, Tooyama I. Dietary Strategies to Mitigate Alzheimer's Disease: Insights into Antioxidant Vitamin Intake and Supplementation with Microbiota-Gut-Brain Axis Cross-Talk. Antioxidants (Basel) 2024; 13:1504. [PMID: 39765832 PMCID: PMC11673287 DOI: 10.3390/antiox13121504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Alzheimer's disease (AD), which is characterized by deterioration in cognitive function and neuronal death, is the most prevalent age-related progressive neurodegenerative disease. Clinical and experimental research has revealed that gut microbiota dysbiosis may be present in AD patients. The changed gut microbiota affects brain function and behavior through several mechanisms, including tau phosphorylation and increased amyloid deposits, neuroinflammation, metabolic abnormalities, and persistent oxidative stress. The lack of effective treatments to halt or reverse the progression of this disease has prompted a search for non-pharmaceutical tools. Modulation of the gut microbiota may be a promising strategy in this regard. This review aims to determine whether specific dietary interventions, particularly antioxidant vitamins, either obtained from the diet or as supplements, may support the formation of beneficial microbiota in order to prevent AD development by contributing to the systemic reduction of chronic inflammation or by acting locally in the gut. Understanding their roles would be beneficial as it may have the potential to be used as a future therapy option for AD patients.
Collapse
Affiliation(s)
- Wan Zurinah Wan Ngah
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| | - Hajar Fauzan Ahmad
- Department of Industrial Biotechnology, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Gambang 26300, Pahang, Malaysia;
| | - Sheril June Ankasha
- Unisza Science and Medicine Foundation Centre, Universiti Sultan Zainal Abidin, Gong Badak Campus, Kuala Nerus 21300, Terengganu, Malaysia;
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Ikuo Tooyama
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| |
Collapse
|
9
|
Dey S, Kumar R, Mishra R, Bera S. Exploring cross-α amyloids: from functional roles to design innovations. Trends Biochem Sci 2024; 49:1097-1110. [PMID: 39510919 DOI: 10.1016/j.tibs.2024.10.004] [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] [Received: 07/15/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024]
Abstract
Amyloids are filamentous protein aggregates that have traditionally been associated with neurodegenerative diseases, although they are also known to play pivotal functional roles across diverse forms of life. Although the cross-β structure has represented the hallmark of amyloidal assemblies, a cross-α structure was recently characterized as a functional microbial amyloid, and further work has shown that de novo designed sequences also assemble into cross-α amyloids, emphasizing cross-α as an alternative paradigm for self-assembly into ordered aggregates. In this review, we summarize recent discoveries of cross-α amyloids both in nature and artificially designed systems, and we describe their fundamental structural organization, self-assembly mechanisms, and biological functions. Finally, we outline the future opportunities for research and development in this potential field.
Collapse
Affiliation(s)
- Sukantha Dey
- Department of Chemistry, Ashoka University, Sonipat, Haryana 131029, India
| | - Rohit Kumar
- Department of Chemistry, Ashoka University, Sonipat, Haryana 131029, India
| | - Rajkumar Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, S.A.S. Nagar (Mohali) 160062, India
| | - Santu Bera
- Department of Chemistry, Ashoka University, Sonipat, Haryana 131029, India.
| |
Collapse
|
10
|
Bigo-Simon A, Estrozi LF, Chaumont A, Schurhammer R, Schoehn G, Combet J, Schmutz M, Schaaf P, Jierry L. 3D Cryo-Electron Microscopy Reveals the Structure of a 3-Fluorenylmethyloxycarbonyl Zipper Motif Ensuring the Self-Assembly of Tripeptide Nanofibers. ACS NANO 2024; 18:30448-30462. [PMID: 39441741 DOI: 10.1021/acsnano.4c08043] [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: 10/25/2024]
Abstract
Short peptide-based supramolecular hydrogels appeared as highly interesting materials for applications in many fields. The optimization of their properties relies mainly on the design of a suitable hydrogelator through an empirical trial-and-error strategy based on the synthesis of various types of peptides. This approach is in part due to the lack of prior structural knowledge of the molecular architecture of the various families of nanofibers. The 3D structure of the nanofibers determines their ability to interact with entities present in their surrounding environment. Thus, it is important to resolve the internal structural organization of the material. Herein, using Fmoc-FFY tripeptide as a model amphiphilic hydrogelator and cryo-EM reconstruction approach, we succeeded to obtain a 3.8 Å resolution 3D structure of a self-assembled nanofiber with a diameter of approximately 4.1 nm and with apparently "infinite" length. The elucidation of the spatial organization of such nano-objects addresses fundamental questions about the way short amphiphilic N-Fmoc peptides lacking secondary structure can self-assemble and ensure the cohesion of such a lengthy nanostructure. This nanofiber is organized into a triple-stranded helix with an asymmetric unit composed of two Fmoc-FFY peptides per strand. The three identical amphiphilic strands are maintained together by strong lateral interactions coming from a 3-Fmoc zipper motif. This hydrophobic core of the nanofiber is surrounded by 12 phenyl groups from phenylalanine residues, nonplanar with the six Fmoc groups. Polar tyrosine residues at the C-term position constitute the hydrophilic shell and are exposed all around the external part of the assembly. This fiber has a highly hydrophobic central core with an internal diameter of only 2.4 Å. Molecular dynamics simulations highlight van der Waals and hydrogen bonds between peptides placed on top of each other. We demonstrate that the self-assembly of Fmoc-FFY, whether induced by annealing or by the action of a phosphatase on the phosphorylated precursor Fmoc-FFpY, results in two nanostructures with minor differences that we are unable to distinguish.
Collapse
Affiliation(s)
- Alexis Bigo-Simon
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
- Faculté de Chimie, Université de Strasbourg, UMR7140, 1 rue Blaise Pascal, Strasbourg Cedex 67008, France
| | - Leandro F Estrozi
- CNRS, CEA, IBSUniversité de Grenoble Alpes, Grenoble F-38000, France
| | - Alain Chaumont
- Faculté de Chimie, Université de Strasbourg, UMR7140, 1 rue Blaise Pascal, Strasbourg Cedex 67008, France
| | - Rachel Schurhammer
- Faculté de Chimie, Université de Strasbourg, UMR7140, 1 rue Blaise Pascal, Strasbourg Cedex 67008, France
| | - Guy Schoehn
- CNRS, CEA, IBSUniversité de Grenoble Alpes, Grenoble F-38000, France
| | - Jérôme Combet
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
| | - Marc Schmutz
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
| | - Pierre Schaaf
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
- INSERM Unite 1121, CRBSInstitut National de la Santé et de la Recherche Médicale, 1 rue Eugène Boeckel, Strasbourg 67000, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 8 rue Sainte Elisabeth, Strasbourg 67000, France
| | - Loïc Jierry
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
| |
Collapse
|
11
|
Patel SP, Nikam T, Sreepathi B, Karankar VS, Jaiswal A, Vardhan SV, Rana A, Toga V, Srivastava N, Saraf SA, Awasthi S. Unraveling the Molecular Jam: How Crowding Shapes Protein Aggregation in Neurodegenerative Disorders. ACS Chem Biol 2024; 19:2118-2130. [PMID: 39373539 DOI: 10.1021/acschembio.4c00365] [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: 10/08/2024]
Abstract
Protein misfolding and aggregation are the hallmarks of neurodegenerative diseases including Huntington's disease, Parkinson's disease, Alzheimer's disease, and prion diseases. A crowded cellular environment plays a crucial role in modulating protein aggregation processes in vivo and the pathological aggregation of proteins linked to different neurodegenerative disorders. Here, we review recent studies examining the effects of various crowding agents, such as polysaccharides, polyethylene glycol, and proteins like BSA and lysozyme on the behaviors of aggregation of several amyloidogenic peptides and proteins, including amylin, huntingtin, tau, α-synuclein, prion, and amyloid-β. We also summarize how the aggregation kinetics, thermodynamic stability, and morphology of amyloid fibrils are altered significantly in the presence of crowding agents. In addition, we also discuss the molecular basis underlying the modulation of amyloidogenic aggregation, focusing on changes in the protein conformation, and the nucleation mechanism. The molecular understanding of the effects of macromolecular crowding on amyloid aggregation is essential for revealing disease pathologies and identifying possible therapeutic targets. Thus, this review offers a perspective on the complex interplay between protein aggregation and the crowded cellular environment in vivo and explains the relevance of crowding in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Shashi Prakash Patel
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Tejas Nikam
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Bhargavi Sreepathi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Vijayshree S Karankar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Ankita Jaiswal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Salumuri Vamsi Vardhan
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Anika Rana
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Vanshu Toga
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Shubhini A Saraf
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Saurabh Awasthi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| |
Collapse
|
12
|
Kell DB, Pretorius E. Proteomic Evidence for Amyloidogenic Cross-Seeding in Fibrinaloid Microclots. Int J Mol Sci 2024; 25:10809. [PMID: 39409138 PMCID: PMC11476703 DOI: 10.3390/ijms251910809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
In classical amyloidoses, amyloid fibres form through the nucleation and accretion of protein monomers, with protofibrils and fibrils exhibiting a cross-β motif of parallel or antiparallel β-sheets oriented perpendicular to the fibre direction. These protofibrils and fibrils can intertwine to form mature amyloid fibres. Similar phenomena can occur in blood from individuals with circulating inflammatory molecules (and also some originating from viruses and bacteria). Such pathological clotting can result in an anomalous amyloid form termed fibrinaloid microclots. Previous proteomic analyses of these microclots have shown the presence of non-fibrin(ogen) proteins, suggesting a more complex mechanism than simple entrapment. We thus provide evidence against such a simple entrapment model, noting that clot pores are too large and centrifugation would have removed weakly bound proteins. Instead, we explore whether co-aggregation into amyloid fibres may involve axial (multiple proteins within the same fibril), lateral (single-protein fibrils contributing to a fibre), or both types of integration. Our analysis of proteomic data from fibrinaloid microclots in different diseases shows no significant quantitative overlap with the normal plasma proteome and no correlation between plasma protein abundance and their presence in fibrinaloid microclots. Notably, abundant plasma proteins like α-2-macroglobulin, fibronectin, and transthyretin are absent from microclots, while less abundant proteins such as adiponectin, periostin, and von Willebrand factor are well represented. Using bioinformatic tools, including AmyloGram and AnuPP, we found that proteins entrapped in fibrinaloid microclots exhibit high amyloidogenic tendencies, suggesting their integration as cross-β elements into amyloid structures. This integration likely contributes to the microclots' resistance to proteolysis. Our findings underscore the role of cross-seeding in fibrinaloid microclot formation and highlight the need for further investigation into their structural properties and implications in thrombotic and amyloid diseases. These insights provide a foundation for developing novel diagnostic and therapeutic strategies targeting amyloidogenic cross-seeding in blood clotting disorders.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Søltofts Plads 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
13
|
Dabas A, Goyal B. Structural Reorganization Mechanism of the Aβ 42 Fibril Mediated by N-Substituted Oligopyrrolamide ADH-353. ACS Chem Neurosci 2024; 15:3136-3151. [PMID: 39158263 DOI: 10.1021/acschemneuro.4c00253] [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: 08/20/2024] Open
Abstract
The inhibition of amyloid-β (Aβ) fibrillation and clearance of Aβ aggregates have emerged as a potential pharmacological strategy to alleviate Aβ aggregate-induced neurotoxicity in Alzheimer's disease (AD). Maity et al. shortlisted ADH-353 from a small library of positively charged N-substituted oligopyrrolamides for its notable ability to inhibit Aβ fibrillation, disintegrate intracellular cytotoxic Aβ oligomers, and alleviate Aβ-induced cytotoxicity in the SH-SY5Y and N2a cells. However, the molecular mechanism through which ADH-353 interacts with the Aβ42 fibrils, leading to their disruption and subsequent clearance, remains unclear. Thus, a detailed molecular mechanism underlying the disruption of neurotoxic Aβ42 fibrils (PDB ID 2NAO) by ADH-353 has been illuminated in this work using molecular dynamics simulations. Interestingly, conformational snapshots during simulation depicted the shortening and disappearance of β-strands and the emergence of a helix conformation, indicating a loss of the well-organized β-sheet-rich structure of the disease-relevant Aβ42 fibril on the incorporation of ADH-353. ADH-353 binds strongly to the Aβ42 fibril (ΔGbinding= -142.91 ± 1.61 kcal/mol) with a notable contribution from the electrostatic interactions between positively charged N-propylamine side chains of ADH-353 with the glutamic (Glu3, Glu11, and Glu22) and aspartic (Asp7 and Asp23) acid residues of the Aβ42 fibril. This aligns well with heteronuclear single quantum coherence NMR studies, which depict that the binding of ADH-353 with the Aβ peptide is driven by electrostatic and hydrophobic contacts. Furthermore, a noteworthy decrease in the binding affinity of Aβ42 fibril chains on the incorporation of ADH-353 indicates the weakening of interchain interactions leading to the disruption of the double-horseshoe conformation of the Aβ42 fibril. The illumination of key interactions responsible for the destabilization of the Aβ42 fibril by ADH-353 in this work will greatly aid in designing new chemical scaffolds with enhanced efficacy for the clearance of Aβ aggregates in AD.
Collapse
Affiliation(s)
- Arushi Dabas
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| | - Bhupesh Goyal
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| |
Collapse
|
14
|
Li J, Zheng Z, Ma Y, Dong Z, Li MH, Hu J. Mechanically Ultra-Robust Fluorescent Elastomer for Elaborating Auxetic Composite. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402130. [PMID: 38678509 DOI: 10.1002/smll.202402130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Fluorescent elastomers are predominantly fabricated through doping fluorescent components or conjugating chromophores into polymer networks, which often involves detrimental effects on mechanical performance and also makes large-scale production difficult. Inspired by the heteroatom-rich microphase separation structures assisted by intensive hydrogen bonds in natural organisms, an ultra-robust fluorescent polyurethane elastomer is reported, which features a remarkable fracture strength of 87.2 MPa with an elongation of 1797%, exceptional toughness of 678.4 MJ m-3 and intrinsic cyan fluorescence at 445 nm. Moreover, the reversible fluorescence variation with temperature could in situ reveal the microphase separation of the elastomer in real time. By taking advantage of mechanical properties, intrinsic fluorescence and hydrogen bonds-promoted interfacial bonding ability, this fluorescent elastomer can be utilized as an auxetic skeleton for the elaboration of an integrated auxetic composite. Compared with the auxetic skeleton alone, the integrated composite shows an improved mechanical performance while maintaining auxetic deformation in a large strain below 185%, and its auxetic process can be visually detected under ultraviolet light by the fluorescence of the auxetic skeleton. The concept of introducing hydrogen-bonded heteroatom-rich microphase separation structures into polymer networks in this work provides a promising approach to developing fluorescent elastomers with exceptional mechanical properties.
Collapse
Affiliation(s)
- Jiawei Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, North Third Ring Road 15, Chaoyang District, Beijing, 100029, China
| | - Zhiran Zheng
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, North Third Ring Road 15, Chaoyang District, Beijing, 100029, China
| | - Yaning Ma
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, North Third Ring Road 15, Chaoyang District, Beijing, 100029, China
| | - Zhaoxing Dong
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, North Third Ring Road 15, Chaoyang District, Beijing, 100029, China
| | - Min-Hui Li
- Chimie ParisTech, PSL University, CNRS, Institut de Recherche de Chimie Paris, 11 rue Pierre et Marie Curie, Paris, 75005, France
| | - Jun Hu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, North Third Ring Road 15, Chaoyang District, Beijing, 100029, China
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Renmin Street 5625, Chaoyang District, Changchun, 130022, China
| |
Collapse
|
15
|
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
|
16
|
Heath SG, Naughton JD, Magon NJ, Gray SG, Smith BR, Morris VK, Göbl C. Characterizing the amyloid core region of the tumor suppressor protein p16 INK4a using a limited proteolysis and peptide-based approach. J Biol Chem 2024; 300:107590. [PMID: 39032649 PMCID: PMC11375262 DOI: 10.1016/j.jbc.2024.107590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024] Open
Abstract
The human tumor suppressor p16INK4a is a small monomeric protein that can form amyloid structures. Formation of p16INK4a amyloid fibrils is induced by oxidation which creates an intermolecular disulfide bond. The conversion into amyloid is associated with a change from an all α-helical structure into β-sheet fibrils. Currently, structural insights into p16INK4a amyloid fibrils are lacking. Here, we investigate the amyloid-forming regions of this tumor suppressor using isotope-labeling limited-digestion mass spectrometry analysis. We discover two key regions that likely form the structured core of the amyloid. Further investigations using thioflavin-T fluorescence assays, electron microscopy, and solution nuclear magnetic resonance spectroscopy of shorter peptide regions confirm the self-assembly of the identified sequences that include methionine and leucine repeat regions. This work describes a simple approach for studying protein motifs involved in the conversion of monomeric species into aggregated fibril structures. It provides insight into the polypeptide sequence underlying the core structure of amyloid p16INK4a formed after a unique oxidation-driven structural transition.
Collapse
Affiliation(s)
- Sarah G Heath
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Jennifer D Naughton
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Nicholas J Magon
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Shelby G Gray
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Briana R Smith
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Vanessa K Morris
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand.
| | - Christoph Göbl
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand.
| |
Collapse
|
17
|
Monteiro C, Gomes MC, Bharmoria P, Freire MG, Coutinho JA, Custódio CA, Mano JF. Human Platelet Lysate-Derived Nanofibrils as Building Blocks to Produce Free-Standing Membranes for Cell Self-Aggregation. ACS NANO 2024; 18:15815-15830. [PMID: 38833572 PMCID: PMC11191744 DOI: 10.1021/acsnano.4c02790] [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: 02/28/2024] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Amyloid-like fibrils are garnering keen interest in biotechnology as supramolecular nanofunctional units to be used as biomimetic platforms to control cell behavior. Recent insights into fibril functionality have highlighted their importance in tissue structure, mechanical properties, and improved cell adhesion, emphasizing the need for scalable and high-kinetics fibril synthesis. In this study, we present the instantaneous and bulk formation of amyloid-like nanofibrils from human platelet lysate (PL) using the ionic liquid cholinium tosylate as a fibrillating agent. The instant fibrillation of PL proteins upon supramolecular protein-ionic liquid interactions was confirmed from the protein conformational transition toward cross-β-sheet-rich structures. These nanofibrils were utilized as building blocks for the formation of thin and flexible free-standing membranes via solvent casting to support cell self-aggregation. These PL-derived fibril membranes reveal a nanotopographically rough surface and high stability over 14 days under cell culture conditions. The culture of mesenchymal stem cells or tumor cells on the top of the membrane demonstrated that cells are able to adhere and self-organize in a three-dimensional (3D) spheroid-like microtissue while tightly folding the fibril membrane. Results suggest that nanofibril membrane incorporation in cell aggregates can improve cell viability and metabolic activity, recreating native tissues' organization. Altogether, these PL-derived nanofibril membranes are suitable bioactive platforms to generate 3D cell-guided microtissues, which can be explored as bottom-up strategies to faithfully emulate native tissues in a fully human microenvironment.
Collapse
Affiliation(s)
- Cátia
F. Monteiro
- CICECO − Aveiro Institute
of Materials, Department of Chemistry, University
of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Maria C. Gomes
- CICECO − Aveiro Institute
of Materials, Department of Chemistry, University
of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | | | - Mara G. Freire
- CICECO − Aveiro Institute
of Materials, Department of Chemistry, University
of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - João A.
P. Coutinho
- CICECO − Aveiro Institute
of Materials, Department of Chemistry, University
of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Catarina A. Custódio
- CICECO − Aveiro Institute
of Materials, Department of Chemistry, University
of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - João F. Mano
- CICECO − Aveiro Institute
of Materials, Department of Chemistry, University
of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| |
Collapse
|
18
|
Farzam F, Dabirmanesh B. Experimental techniques for detecting and evaluating the amyloid fibrils. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 206:183-227. [PMID: 38811081 DOI: 10.1016/bs.pmbts.2024.03.004] [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: 05/31/2024]
Abstract
Amyloid fibrils are insoluble proteins with intricate β-sheet structures associated with various human diseases, including Parkinson's, Alzheimer's, and prion diseases. Proteins can form aggregates when their structure is misfolded, resulting in highly organized amyloid fibrils or amorphous aggregates. The formation of protein aggregates is a promising research field for mitigating diseases and the pharmaceutical and food industries. It is important to monitor and minimize the appearance of aggregates in these protein products. Several methods exist to assess protein aggregation, that includes from basic investigations to advanced biophysical techniques. Physicochemical parameters such as molecular weight, conformation, structure, and dimension are examined to study aggregation. There is an urgent need to develop methods for the detection of protein aggregation and amyloid fibril formation both in vitro and in vivo. This chapter focuses on a comprehensive discussion of the methods used to characterize and evaluate aggregates and amyloid fibrils.
Collapse
Affiliation(s)
- Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
19
|
Duran-Meza E, Araya-Secchi R, Romero-Hasler P, Soto-Bustamante EA, Castro-Fernandez V, Castillo-Caceres C, Monasterio O, Diaz-Espinoza R. Metal Ions Can Modulate the Self-Assembly and Activity of Catalytic Peptide Amyloids. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:6094-6106. [PMID: 38470353 DOI: 10.1021/acs.langmuir.3c02983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Rational design of peptides has become a powerful tool to produce self-assembled nanostructures with the ability to catalyze different chemical reactions, paving the way to develop minimalistic enzyme-like nanomaterials. Catalytic amyloid-like assemblies have emerged among the most versatile and active, but they often require additional factors for activity. Elucidating how these factors influence the structure and activity is key for the design. Here, we showed that biologically relevant metal ions can guide and modulate the self-assembly of a small peptide into diverse amyloid architectures. The morphology and catalytic activity of the resulting fibrils were tuned by the specific metal ion decorating the surface, whereas X-ray structural analysis of the amyloids showed ion-dependent shape sizes. Molecular dynamics simulations showed that the metals can strongly affect the local conformational space, which can trigger major rearrangements of the fibrils. Our results demonstrate that the conformational landscape of catalytic amyloids is broad and tunable by external factors, which can be critical for future design strategies.
Collapse
Affiliation(s)
- Eva Duran-Meza
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, General Amengual 014, Estación Central, Santiago 9170390, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile
| | - Raul Araya-Secchi
- Computational Biophysics group, Facultad de Ingenieria, Tecnologia y Diseño, Universidad San Sebastian, Bellavista 7, Recoleta, Santiago 8420524, Chile
- Centro Basal Ciencia & Vida, Universidad San Sebastian, Santiago 8420524, Chile
| | - Patricio Romero-Hasler
- Departamento de Ciencia de los Alimentos y Tecnología Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, Santiago 81380494, Chile
| | - Eduardo Arturo Soto-Bustamante
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, Santiago 81380494, Chile
| | - Victor Castro-Fernandez
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile
| | - Claudio Castillo-Caceres
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, General Amengual 014, Estación Central, Santiago 9170390, Chile
| | - Octavio Monasterio
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile
| | - Rodrigo Diaz-Espinoza
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, General Amengual 014, Estación Central, Santiago 9170390, Chile
| |
Collapse
|
20
|
Song Z, Li Y, Li R, Fan GC, Luo X. Robust Electrochemical Biosensors Based on Antifouling Peptide Nanoparticles for Protein Quantification in Complex Biofluids. ACS Sens 2024; 9:1525-1532. [PMID: 38377562 DOI: 10.1021/acssensors.3c02706] [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: 02/22/2024]
Abstract
Peptides with distinct physiochemical properties and biocompatibility hold significant promise across diverse domains including antifouling biosensors. However, the stability of natural antifouling peptides in physiological conditions poses significant challenges to their viability for sustained practical applications. Herein, a unique antifouling peptide FFFGGGEKEKEKEK was designed and self-assembled to form peptide nanoparticles (PNPs), which possessed enhanced stability against enzymatic hydrolysis in biological fluids. The PNP-coated interfaces exhibited superior stability and antifouling properties in preventing adsorption of nonspecific materials, such as proteins and cells in biological samples. Moreover, a highly sensitive and ultralow fouling electrochemical biosensor was developed through the immobilization of the PNPs and specific aptamers onto the polyaniline nanowire-modified electrode, achieving the biomarker carcinoembryonic antigen detection in complex biofluids with reliable accuracy. This research not only addresses the challenge of the poor proteolytic resistance observed in natural peptides but also introduces a universal strategy for constructing ultralow fouling sensing devices.
Collapse
Affiliation(s)
- Zhen Song
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yang Li
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Rong Li
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Gao-Chao Fan
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
21
|
Lee D, Jung HG, Park D, Bang J, Cheong DY, Jang JW, Kim Y, Lee S, Lee SW, Lee G, Kim YH, Hong JH, Hwang KS, Lee JH, Yoon DS. Bioengineered amyloid peptide for rapid screening of inhibitors against main protease of SARS-CoV-2. Nat Commun 2024; 15:2108. [PMID: 38453923 PMCID: PMC10920794 DOI: 10.1038/s41467-024-46296-7] [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: 05/13/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has evoked a worldwide pandemic. As the emergence of variants has hampered the neutralization capacity of currently available vaccines, developing effective antiviral therapeutics against SARS-CoV-2 and its variants becomes a significant challenge. The main protease (Mpro) of SARS-CoV-2 has received increased attention as an attractive pharmaceutical target because of its pivotal role in viral replication and proliferation. Here, we generated a de novo Mpro-inhibitor screening platform to evaluate the efficacies of Mpro inhibitors based on Mpro cleavage site-embedded amyloid peptide (MCAP)-coated gold nanoparticles (MCAP-AuNPs). We fabricated MCAPs comprising an amyloid-forming sequence and Mpro-cleavage sequence, mimicking in vivo viral replication process mediated by Mpro. By measuring the proteolytic activity of Mpro and the inhibitory efficacies of various drugs, we confirmed that the MCAP-AuNP-based platform was suitable for rapid screening potential of Mpro inhibitors. These results demonstrated that our MCAP-AuNP-based platform has great potential for discovering Mpro inhibitors and may accelerate the development of therapeutics against COVID-19.
Collapse
Affiliation(s)
- Dongtak Lee
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Hyo Gi Jung
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea
| | - Dongsung Park
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Junho Bang
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea
| | - Da Yeon Cheong
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, 30019, South Korea
- Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Korea University, Sejong, 30019, South Korea
| | - Jae Won Jang
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea
| | - Yonghwan Kim
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea
| | - Seungmin Lee
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Department of Electrical Engineering, Kwangwoon University, Seoul, 01897, South Korea
| | - Sang Won Lee
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Gyudo Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, 30019, South Korea
- Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Korea University, Sejong, 30019, South Korea
| | - Yeon Ho Kim
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea
| | - Ji Hye Hong
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea
- Department of Electrical Engineering, Kwangwoon University, Seoul, 01897, South Korea
| | - Kyo Seon Hwang
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, 02447, South Korea.
| | - Jeong Hoon Lee
- Department of Electrical Engineering, Kwangwoon University, Seoul, 01897, South Korea.
| | - Dae Sung Yoon
- School of Biomedical Engineering, Korea University, Seoul, 02841, South Korea.
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea.
- Astrion Inc, Seoul, 02841, South Korea.
| |
Collapse
|
22
|
Baek Y, Lee M. Solid-state NMR spectroscopic analysis for structure determination of a zinc-bound catalytic amyloid fibril. Methods Enzymol 2024; 697:435-471. [PMID: 38816132 DOI: 10.1016/bs.mie.2024.01.025] [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
Zinc ions are commonly involved in enzyme catalysis and protein structure stabilization, but their coordination geometry of zinc-protein complex is rarely determined. Here, in this chapter, we introduce a systematic solid-state NMR approach to determine the oligomeric assembly and Zn2+ coordination geometry of a de novo designed amyloid fibrils that catalyze zinc dependent ester hydrolysis. NMR chemical shifts and intermolecular contacts confirm that the peptide forms parallel-in-register β-sheets, with the two forms of Zn2+ bound histidines in each peptide. The amphiphilic parallel β-sheets assemble into stacked bilayers that are stabilized by hydrophobic side chains between β-sheets. The conformations of the histidine side chains, determined by 13C-15N distance measurements, reveal how histidines protrude from the β-sheet. 1H-15N correlation spectra show that the single-Zn2+ coordinated histidine associated with dynamic water. The resulting structure provides insight into how metal ions contribute to stabilizing the protein structure and driving its catalytic reactivity.
Collapse
Affiliation(s)
- Yoongyeong Baek
- Department of Chemistry, Drexel University, Philadelphia, PA, United States
| | - Myungwoon Lee
- Department of Chemistry, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Kaygisiz K, Rauch‐Wirth L, Iscen A, Hartenfels J, Kremer K, Münch J, Synatschke CV, Weil T. Peptide Amphiphiles as Biodegradable Adjuvants for Efficient Retroviral Gene Delivery. Adv Healthc Mater 2024; 13:e2301364. [PMID: 37947246 PMCID: PMC11468294 DOI: 10.1002/adhm.202301364] [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: 04/28/2023] [Revised: 10/20/2023] [Indexed: 11/12/2023]
Abstract
Retroviral gene delivery is the key technique for in vitro and ex vivo gene therapy. However, inefficient virion-cell attachment resulting in low gene transduction efficacy remains a major challenge in clinical applications. Adjuvants for ex vivo therapy settings need to increase transduction efficiency while being easily removed or degraded post-transduction to prevent the risk of venous embolism after infusing the transduced cells back to the bloodstream of patients, yet no such peptide system have been reported thus far. In this study, peptide amphiphiles (PAs) with a hydrophobic fatty acid and a hydrophilic peptide moiety that reveal enhanced viral transduction efficiency are introduced. The PAs form β-sheet-rich fibrils that assemble into positively charged aggregates, promoting virus adhesion to the cell membrane. The block-type amphiphilic sequence arrangement in the PAs ensures efficient cell-virus interaction and biodegradability. Good biodegradability is observed for fibrils forming small aggregates and it is shown that via molecular dynamics simulations, the fibril-fibril interactions of PAs are governed by fibril surface hydrophobicity. These findings establish PAs as additives in retroviral gene transfer, rivalling commercially available transduction enhancers in efficiency and degradability with promising translational options in clinical gene therapy applications.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Lena Rauch‐Wirth
- Institute of Molecular VirologyUlm University Medical CenterMeyerhofstraße 189081UlmGermany
| | - Aysenur Iscen
- Polymer Theory DepartmentMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jan Hartenfels
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Kurt Kremer
- Polymer Theory DepartmentMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jan Münch
- Institute of Molecular VirologyUlm University Medical CenterMeyerhofstraße 189081UlmGermany
| | - Christopher V. Synatschke
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Tanja Weil
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| |
Collapse
|
24
|
Hosseini AN, van der Spoel D. Martini on the Rocks: Can a Coarse-Grained Force Field Model Crystals? J Phys Chem Lett 2024; 15:1079-1088. [PMID: 38261634 PMCID: PMC10839907 DOI: 10.1021/acs.jpclett.4c00012] [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: 01/02/2024] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Computational chemistry is an important tool in numerous scientific disciplines, including drug discovery and structural biology. Coarse-grained models offer simple representations of molecular systems that enable simulations of large-scale systems. Because there has been an increase in the adoption of such models for simulations of biomolecular systems, critical evaluation is warranted. Here, the stability of the amyloid peptide and organic crystals is evaluated using the Martini 3 coarse-grained force field. The crystals change shape drastically during the simulations. Radial distribution functions show that the distance between backbone beads in β-sheets increases by ∼1 Å, breaking the crystals. The melting points of organic compounds are much too low in the Martini force field. This suggests that Martini 3 lacks the specific interactions needed to accurately simulate peptides or organic crystals without imposing artificial restraints. The problems may be exacerbated by the use of the 12-6 potential, suggesting that a softer potential could improve this model for crystal simulations.
Collapse
Affiliation(s)
- A. Najla Hosseini
- Department of Cell and Molecular
Biology, Uppsala University, Box 596, SE-75124 Uppsala, Sweden
| | - David van der Spoel
- Department of Cell and Molecular
Biology, Uppsala University, Box 596, SE-75124 Uppsala, Sweden
| |
Collapse
|
25
|
Samui S, Biswas S, Basak S, Ghosh S, Muniyappa K, Naskar J. De novo designed aliphatic and aromatic peptides assemble into amyloid-like cytotoxic supramolecular nanofibrils. RSC Adv 2024; 14:4382-4388. [PMID: 38304566 PMCID: PMC10831423 DOI: 10.1039/d3ra07869h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
Peptides are very interesting biomolecules that upon self-association form a variety of thermodynamically stable supramolecular structures of nanometric dimension e.g. nanotubes, nanorods, nanovesicles, nanofibrils, nanowires and many others. Herein, we report six peptide molecules having a general chemical structure, H-Gaba-X-X-OH (Gaba: γ-aminobutyric acid, X: amino acid). Out of these six peptides, three are aromatic and the others are aliphatic. Atomic force microscopic (AFM) studies reveal that except peptide 6 (H-Gaba-Trp-Trp-OH), all the reported peptides adopt nanofibrillar morphology upon aggregation in aqueous medium. These supramolecular assemblies can recognize amyloid-specific molecular probe congo red (CR) and thioflavine t (ThT) and exhibit all the characteristic properties of amyloids. The MTT cell viability assay reveals that the toxicity of both aliphatic and aromatic peptides increases with increasing concentration of the peptides to both cancer (HeLa) and non-cancer (HEK 293) cells. Of note, the aromatic peptides show a slightly higher cytotoxic effect compared to the aliphatic peptides. Overall, the studies highlight the self-assembling nature of the de novo designed aliphatic and aromatic peptides and pave the way towards elucidating the intricacies of pathogenic amyloid assemblies.
Collapse
Affiliation(s)
- Satyabrata Samui
- Department of Biochemistry and Biophysics, University of Kalyani Nadia WB 741235 India
| | - Soumi Biswas
- Department of Biochemistry and Biophysics, University of Kalyani Nadia WB 741235 India
| | - Shubhanwita Basak
- Department of Biochemistry and Biophysics, University of Kalyani Nadia WB 741235 India
| | - Shreya Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani Nadia WB 741235 India
| | - K Muniyappa
- Department of Biochemistry, Indian Institute of Science Bangalore Karnataka 560 012 India
| | - Jishu Naskar
- Department of Biochemistry and Biophysics, University of Kalyani Nadia WB 741235 India
| |
Collapse
|
26
|
Prasanna AM, Sen P. Recent Developments of Hybrid Fluorescence Techniques: Advances in Amyloid Detection Methods. Curr Protein Pept Sci 2024; 25:667-681. [PMID: 38715332 DOI: 10.2174/0113892037291597240429094515] [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: 12/26/2023] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 09/21/2024]
Abstract
Amyloid fibrils are formed from various pathological proteins. Monitoring their aggregation process is necessary for early detection and treatment. Among the available detection techniques, fluorescence is simple, intuitive, and convenient due to its sensitive and selective mode of detection. It has certain disadvantages like poor photothermal stability and detection state limitation. Research has focused on minimising the limitation by developing hybrid fluorescence techniques. This review focuses on the two ways fluorescence (intrinsic and extrinsic) has been used to monitor amyloid fibrils. In intrinsic/label free fluorescence: i) The fluorescence emission through aromatic amino acid residues like phenylalanine (F), tyrosine (Y) and tryptophan (W) is present in amyloidogenic peptides/protein sequence. And ii) The structural changes from alpha helix to cross-β-sheet structures during amyloid formation contribute to the fluorescence emission. The second method focuses on the use of extrinsic fluorophores to monitor amyloid fibrils i) organic dyes/small molecules, ii) fluorescent tagged proteins, iii) nanoparticles, iv) metal complexes and v) conjugated polymers. All these fluorophores have their own limitations. Developing them into hybrid fluorescence techniques and converting it into biosensors can contribute to early detection of disease.
Collapse
Affiliation(s)
- A Miraclin Prasanna
- Centre for Bio Separation Technology (CBST), School of Biosciences and Technology, VIT, Vellore, 632014, Tamil Nadu, India
| | - Priyankar Sen
- Centre for Bio Separation Technology (CBST), School of Biosciences and Technology, VIT, Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
27
|
Zhang H, Lv S, Jin C, Ren F, Wang J. Wheat gluten amyloid fibrils: Conditions, mechanism, characterization, application, and future perspectives. Int J Biol Macromol 2023; 253:126435. [PMID: 37611682 DOI: 10.1016/j.ijbiomac.2023.126435] [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: 04/18/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Amyloid fibrils have excellent structural characteristics, such as a high aspect ratio, excellent stiffness, and a wide availability of functional groups on the surface. More studies are now focusing on the formation of amyloid fibrils using food proteins. Protein fibrillation is now becoming recognized as a promising strategy for enhancing the function of food proteins and expanding their range of applications. Wheat gluten is rich in glutamine (Q), hydrophobic amino acids, and the α-helix structure with high β-sheet tendency. These characteristics make it very easy for wheat gluten to form amyloid fibrils. The conditions, formation mechanism, characterization methods, and application of amyloid fibrils formed by wheat gluten are summarized in this review. Further exploration of amyloid fibrils formed by wheat gluten will reveal how they can play a significant role in food, biology, and other fields, especially in medicine.
Collapse
Affiliation(s)
- Huijuan Zhang
- School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Shihao Lv
- School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Chengming Jin
- School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Feiyue Ren
- School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Jing Wang
- School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| |
Collapse
|
28
|
Katina N, Marchenkov V, Ryabova N, Ilyina N, Marchenko N, Balobanov V, Finkelstein A. Influence of Amino Acid Substitutions in ApoMb on Different Stages of Unfolding of Amyloids. Molecules 2023; 28:7736. [PMID: 38067466 PMCID: PMC10707739 DOI: 10.3390/molecules28237736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
To date, most research on amyloid aggregation has focused on describing the structure of amyloids and the kinetics of their formation, while the conformational stability of fibrils remains insufficiently explored. The aim of this work was to investigate the effect of amino acid substitutions on the stability of apomyoglobin (ApoMb) amyloids. A study of the amyloid unfolding of ApoMb and its six mutant variants by urea has been carried out. Changes in the structural features of aggregates during unfolding were recorded by far-UV CD and native electrophoresis. It was shown that during the initial stage of denaturation, amyloids' secondary structure partially unfolds. Then, the fibrils undergo dissociation and form intermediate aggregates weighing approximately 1 MDa, which at the last stage of unfolding decompose into 18 kDa monomeric unfolded molecules. The results of unfolding transitions suggest that the stability of the studied amyloids relative to the intermediate aggregates and of the latter relative to unfolded monomers is higher for ApoMb variants with substitutions that increase the hydrophobicity of the residues. The results presented provide a new insight into the mechanism of stabilization of protein aggregates and can serve as a base for further investigations of the amyloids' stability.
Collapse
Affiliation(s)
- Natalya Katina
- Institute of Protein Research RAS, 142290 Pushchino, Russia; (V.M.); (N.R.); (N.I.); (N.M.)
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 142290 Pushchino, Russia
| | - Victor Marchenkov
- Institute of Protein Research RAS, 142290 Pushchino, Russia; (V.M.); (N.R.); (N.I.); (N.M.)
| | - Natalya Ryabova
- Institute of Protein Research RAS, 142290 Pushchino, Russia; (V.M.); (N.R.); (N.I.); (N.M.)
| | - Nelly Ilyina
- Institute of Protein Research RAS, 142290 Pushchino, Russia; (V.M.); (N.R.); (N.I.); (N.M.)
| | - Natalia Marchenko
- Institute of Protein Research RAS, 142290 Pushchino, Russia; (V.M.); (N.R.); (N.I.); (N.M.)
| | - Vitalii Balobanov
- Institute of Protein Research RAS, 142290 Pushchino, Russia; (V.M.); (N.R.); (N.I.); (N.M.)
| | - Alexey Finkelstein
- Institute of Protein Research RAS, 142290 Pushchino, Russia; (V.M.); (N.R.); (N.I.); (N.M.)
| |
Collapse
|
29
|
Siposova K, Huntosova V, Sedlakova D, Macajova M, Bilcik B, Nair AV, Nair S, Hovhannisyan V, Chen SJ, Musatov A. Biocompatible zeolite-dye composites with anti-amyloidogenic properties. Int J Biol Macromol 2023; 251:126331. [PMID: 37579899 DOI: 10.1016/j.ijbiomac.2023.126331] [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: 03/23/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
One of the most attractive approaches in biomedicine and pharmacy is the application of multifunctional materials. The mesoporous structure of clinoptilolite (CZ) absorbs various types of substances and can be used as a model for studying the carriers for targeted drug delivery with controlled release. CZ-dye composites are fabricated by incorporation into clinoptilolite pores commonly used dyes, aluminum phthalocyanine, zinc porphine, and hypericin. We examined and compared the effect of pure dyes and CZ-dye composites on insulin amyloidogenesis. The formation of insulin amyloid fibrils and the disassembly of preformed fibrils is significantly affected by any of the three compounds, however, the strongest effect is observed for aluminum phthalocyanine indicating a structurally-dependent anti-amyloidogenic activity of the dyes. The incorporation of dyes into CZ particles resulted in enhanced anti-amyloidogenic activity in comparison to pure CZ particles. The cell metabolic activity, biocompatibility and fluorescence biodistribution of the dyes entrapped in the composites were tested in vitro (U87 MG cells) and in vivo in the quail chorioallantoic membrane model. Considering the photoactive properties of the dyes used, we assume their applicability in photodiagnostics and photodynamic therapy. It can also be expected that their anti-amyloidogenic potential can be enhanced by photodynamic effect.
Collapse
Affiliation(s)
- Katarina Siposova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 04001, Kosice, Slovakia; College of Photonics, National Yang Ming Chiao Tung University, Tainan 711, Taiwan.
| | - Veronika Huntosova
- Center for Interdisciplinary Biosciences, Technology, and Innovation Park, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia.
| | - Dagmar Sedlakova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 04001, Kosice, Slovakia.
| | - Mariana Macajova
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska cesta 9, 840 05 Bratislava, Slovakia.
| | - Boris Bilcik
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska cesta 9, 840 05 Bratislava, Slovakia.
| | | | - Sumesh Nair
- College of Photonics, National Yang Ming Chiao Tung University, Tainan 711, Taiwan
| | | | - Shean-Jen Chen
- College of Photonics, National Yang Ming Chiao Tung University, Tainan 711, Taiwan.
| | - Andrey Musatov
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 04001, Kosice, Slovakia.
| |
Collapse
|
30
|
Rajewski BH, Makwana KM, Angera IJ, Geremia DK, Zepeda AR, Hallinan GI, Vidal R, Ghetti B, Serrano AL, Del Valle JR. β-Bracelets: Macrocyclic Cross-β Epitope Mimics Based on a Tau Conformational Strain. J Am Chem Soc 2023; 145:23131-23142. [PMID: 37844142 PMCID: PMC10823581 DOI: 10.1021/jacs.3c06830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
The aggregation of misfolded tau into neurotoxic fibrils is linked to the progression of Alzheimer's disease (AD) and related tauopathies. Disease-associated conformations of filamentous tau are characterized by hydrophobic interactions between side chains on unique and distant β-strand modules within each protomer. Here, we report the design and diversity-oriented synthesis of β-arch peptide macrocycles composed of the aggregation-prone PHF6 hexapeptide of tau and the cross-β module specific to the AD tau fold. Termed "β-bracelets", these proteomimetics assemble in a sequence- and macrocycle-dependent fashion, resulting in amyloid-like fibrils that feature in-register parallel β-sheet structure. Backbone N-amination of a selected β-bracelet affords soluble inhibitors of tau aggregation. We further demonstrate that the N-aminated macrocycles block the prion-like cellular seeding activity of recombinant tau as well as mature fibrils from AD patient extracts. These studies establish β-bracelets as a new class of cross-β epitope mimics and demonstrate their utility in the rational design of molecules targeting amyloid propagation and seeding.
Collapse
Affiliation(s)
- Benjamin H. Rajewski
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Kamlesh M. Makwana
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Isaac J. Angera
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Danielle K. Geremia
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Anna R. Zepeda
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Grace I. Hallinan
- Department of Pathology & Laboratory Medicine and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, 46202, United States
| | - Ruben Vidal
- Department of Pathology & Laboratory Medicine and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, 46202, United States
| | - Bernardino Ghetti
- Department of Pathology & Laboratory Medicine and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, 46202, United States
| | - Arnaldo L. Serrano
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Juan R. Del Valle
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
31
|
Berbon M, Martinez D, Morvan E, Grélard A, Kauffmann B, Waeytens J, Wien F, Arluison V, Habenstein B. Hfq C-terminal region forms a β-rich amyloid-like motif without perturbing the N-terminal Sm-like structure. Commun Biol 2023; 6:1075. [PMID: 37865695 PMCID: PMC10590398 DOI: 10.1038/s42003-023-05462-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023] Open
Abstract
Hfq is a pleitropic actor that serves as stress response and virulence factor in the bacterial cell. To execute its multiple functions, Hfq assembles into symmetric torus-shaped hexamers. Extending outward from the hexameric core, Hfq presents a C-terminal region, described as intrinsically disordered in solution. Many aspects of the role and the structure of this region remain unclear. For instance, in its truncated form it can promote amyloid-like filament assembly. Here, we show that a minimal 11-residue motif at the C-terminal end of Hfq assembles into filaments with amyloid characteristics. Our data suggest that the full-length Hfq in its filamentous state contains a similar molecular fingerprint than that of the short β-strand peptide, and that the Sm-core structure is not affected by filament formation. Hfq proteins might thus co-exist in two forms in vivo, either as isolated, soluble hexamers or as self-assembled hexamers through amyloid-reminiscent interactions, modulating Hfq cellular functions.
Collapse
Affiliation(s)
- Mélanie Berbon
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, Pessac, France
| | - Denis Martinez
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, Pessac, France
| | - Estelle Morvan
- Univ. Bordeaux, CNRS, INSERM, IECB, UAR 3033, Pessac, France
| | - Axelle Grélard
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, Pessac, France
| | - Brice Kauffmann
- Univ. Bordeaux, CNRS, INSERM, IECB, UAR 3033, Pessac, France
| | - Jehan Waeytens
- Structure et Fonction des Membranes Biologiques, Université libre de Bruxelles, Bruxelles, Belgique
| | - Frank Wien
- Synchrotron SOLEIL, L'Orme des Merisiers, Saint Aubin BP48, 91192, Gif-sur-Yvette, France
| | - Véronique Arluison
- Laboratoire Léon Brillouin LLB, UMR12 CEA CNRS, CEA Saclay, 91191, Gif-sur-Yvette, France.
- Université de Paris Cité, UFR SDV, 75013, Paris, France.
| | - Birgit Habenstein
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, Pessac, France.
| |
Collapse
|
32
|
Tan FHP, Najimudin N, Watanabe N, Shamsuddin S, Azzam G. p-Coumaric acid attenuates the effects of Aβ42 in vitro and in a Drosophila Alzheimer's disease model. Behav Brain Res 2023; 452:114568. [PMID: 37414223 DOI: 10.1016/j.bbr.2023.114568] [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: 03/22/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative condition in civilizations worldwide. The distinctive occurrence of amyloid-beta (Aβ) accumulation into insoluble fibrils is part of the disease pathophysiology with Aβ42 being the most toxic and aggressive Aβ species. The polyphenol, p-Coumaric acid (pCA), has been known to boost a number of therapeutic benefits. Here, pCA's potential to counteract the negative effects of Aβ42 was investigated. First, pCA was confirmed to reduce Aβ42 fibrillation using an in vitro activity assay. The compound was next examined on Aβ42-exposed PC12 neuronal cells and was found to significantly decrease Aβ42-induced cell mortality. pCA was then examined using an AD Drosophila melanogaster model. Feeding of pCA partially reversed the rough eye phenotype, significantly lengthened AD Drosophila's lifespan, and significantly enhanced the majority of the AD Drosophila's mobility in a sex-dependent manner. The findings of this study suggest that pCA may have therapeutic benefits for AD.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia.
| | - Nazalan Najimudin
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Nobumoto Watanabe
- USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia; Bioprobe Application Research Unit, RIKEN Centre for Sustainable Resource Science, RIKEN, Japan; Chemical Resource Development Research Unit, RIKEN CSRS, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia; Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Ghows Azzam
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Penang, Malaysia; Malaysia Genome and Vaccine Institute (MGVI), National Institutes of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000 Kajang, Selangor, Malaysia.
| |
Collapse
|
33
|
Wales DJ. Energy Landscapes and Heat Capacity Signatures for Monomers and Dimers of Amyloid-Forming Hexapeptides. Int J Mol Sci 2023; 24:10613. [PMID: 37445791 DOI: 10.3390/ijms241310613] [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: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Amyloid formation is a hallmark of various neurodegenerative disorders. In this contribution, energy landscapes are explored for various hexapeptides that are known to form amyloids. Heat capacity (CV) analysis at low temperature for these hexapeptides reveals that the low energy structures contributing to the first heat capacity feature above a threshold temperature exhibit a variety of backbone conformations for amyloid-forming monomers. The corresponding control sequences do not exhibit such structural polymorphism, as diagnosed via end-to-end distance and a dihedral angle defined for the monomer. A similar heat capacity analysis for dimer conformations obtained using basin-hopping global optimisation shows clear features in end-to-end distance versus dihedral correlation plots, where amyloid-forming sequences exhibit a preference for larger end-to-end distances and larger positive dihedrals. These results hold true for sequences taken from tau, amylin, insulin A chain, a de novo designed peptide, and various control sequences. While there is a little overall correlation between the aggregation propensity and the temperature at which the low-temperature CV feature occurs, further analysis suggests that the amyloid-forming sequences exhibit the key CV feature at a lower temperature compared to control sequences derived from the same protein.
Collapse
Affiliation(s)
- David J Wales
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| |
Collapse
|
34
|
Gupta S, Dasmahapatra AK. Lycopene destabilizes preformed Aβ fibrils: Mechanistic insights from all-atom molecular dynamics simulation. Comput Biol Chem 2023; 105:107903. [PMID: 37320982 DOI: 10.1016/j.compbiolchem.2023.107903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
The therapeutic strategy employing destabilization of the preformed Aβ fibril by various natural compounds, as studied by experimental and computational methods, has been reported significant in curing Alzheimer's disease (AD). However, lycopene (a carotenoid), from terpenes family, needs investigation for its destabilization potential of Aβ fibril. The highest antioxidant potential and ability to cross blood brain barrier makes lycopene a preferred choice as drug lead for treating AD. The current study focuses on investigating the destabilization potential and underpinning mechanism of lycopene on different polymorphic forms of Aβ fibril via Molecular Dynamics (MD) simulation. The key findings highlight binding of lycopene to the outer surface of the chain F of the fibril (2NAO). Herein G9, K16 and V18 residues were found to be involved in van der Waals with the methyl groups of the lycopene. Additionally, Y10 and F20 residues were observed to interact via π-π interactions with CC bonds of the lycopene. The surface mediated binding of lycopene to the fibril is attributed to the large size and structural rigidity of lycopene along with the bulky size of 2NAO and narrow space of fibrillar cavity. The destabilization of the fibril is evident by breakage of inherent H-bonds and hydrophobic interactions in the presence of one lycopene molecule. The lesser β-sheet content explains disorganization of the fibril and bars the higher order aggregation curbing neurotoxicity of the fibril. The higher concentration of the lycopene is not found to be linearly correlated with the extent of destabilization of the fibril. Lycopene is also observed to destabilize the other polymorphic form of Aβ fibril (2BEG), by accessing the fibrillar cavity and lowering the β-sheet content. The destabilization observed by lycopene on two major polymorphs of Aβ fibril explains its potency towards developing an effective therapeutic approach in treating AD.
Collapse
Affiliation(s)
- Shivani Gupta
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Ashok Kumar Dasmahapatra
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India; Center for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
35
|
Björk L, Klingstedt T, Nilsson KPR. Thiophene-Based Ligands: Design, Synthesis and Their Utilization for Optical Assignment of Polymorphic-Disease-Associated Protein Aggregates. Chembiochem 2023; 24:e202300044. [PMID: 36891883 PMCID: PMC10404026 DOI: 10.1002/cbic.202300044] [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: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/10/2023]
Abstract
The development of ligands for detecting protein aggregates is of great interest, as these aggregated proteinaceous species are the pathological hallmarks of several devastating diseases, including Alzheimer's disease. In this regard, thiophene-based ligands have emerged as powerful tools for fluorescent assessment of these pathological entities. The intrinsic conformationally sensitive photophysical properties of poly- and oligothiophenes have allowed optical assignment of disease-associated protein aggregates in tissue sections, as well as real-time in vivo imaging of protein deposits. Herein, we recount the chemical evolution of different generations of thiophene-based ligands, and exemplify their use for the optical distinction of polymorphic protein aggregates. Furthermore, the chemical determinants for achieving a superior fluorescent thiophene-based ligand, as well as the next generation of thiophene-based ligands targeting distinct aggregated species are described. Finally, the directions for future research into the chemical design of thiophene-based ligands that can aid in resolving the scientific challenges around protein aggregation diseases are discussed.
Collapse
Affiliation(s)
- Linnea Björk
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| |
Collapse
|
36
|
Meng R, Zhu H, Deng P, Li M, Ji Q, He H, Jin L, Wang B. Research progress on albumin-based hydrogels: Properties, preparation methods, types and its application for antitumor-drug delivery and tissue engineering. Front Bioeng Biotechnol 2023; 11:1137145. [PMID: 37113668 PMCID: PMC10127125 DOI: 10.3389/fbioe.2023.1137145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Albumin is derived from blood plasma and is the most abundant protein in blood plasma, which has good mechanical properties, biocompatibility and degradability, so albumin is an ideal biomaterial for biomedical applications, and drug-carriers based on albumin can better reduce the cytotoxicity of drug. Currently, there are numerous reviews summarizing the research progress on drug-loaded albumin molecules or nanoparticles. In comparison, the study of albumin-based hydrogels is a relatively small area of research, and few articles have systematically summarized the research progress of albumin-based hydrogels, especially for drug delivery and tissue engineering. Thus, this review summarizes the functional features and preparation methods of albumin-based hydrogels, different types of albumin-based hydrogels and their applications in antitumor drugs, tissue regeneration engineering, etc. Also, potential directions for future research on albumin-based hydrogels are discussed.
Collapse
Affiliation(s)
- Run Meng
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Huimin Zhu
- Sheyang County Comprehensive Inspection and Testing Center, Yancheng, China
| | - Peiying Deng
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Minghui Li
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Qingzhi Ji
- School of Pharmacy, Yancheng Teachers’ University, Yancheng, China
| | - Hao He
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Department of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
37
|
Sun Z, He Q, Gong Z, Kalhor P, Huai Z, Liu Z. A General Picture of Cucurbit[8]uril Host–Guest Binding: Recalibrating Bonded Interactions. Molecules 2023; 28:molecules28073124. [PMID: 37049887 PMCID: PMC10095826 DOI: 10.3390/molecules28073124] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/15/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
Atomic-level understanding of the dynamic feature of host–guest interactions remains a central challenge in supramolecular chemistry. The remarkable guest binding behavior of the Cucurbiturils family of supramolecular containers makes them promising drug carriers. Among Cucurbit[n]urils, Cucurbit[8]uril (CB8) has an intermediate portal size and cavity volume. It can exploit almost all host–guest recognition motifs formed by this host family. In our previous work, an extensive computational investigation of the binding of seven commonly abused and structurally diverse drugs to the CB8 host was performed, and a general dynamic binding picture of CB8-guest interactions was obtained. Further, two widely used fixed-charge models for drug-like molecules were investigated and compared in great detail, aiming at providing guidelines in choosing an appropriate charge scheme in host-guest modelling. Iterative refitting of atomic charges leads to improved binding thermodynamics and the best root-mean-squared deviation from the experimental reference is 2.6 kcal/mol. In this work, we focus on a thorough evaluation of the remaining parts of classical force fields, i.e., the bonded interactions. The widely used general Amber force fields are assessed and refitted with generalized force-matching to improve the intra-molecular conformational preference, and thus the description of inter-molecular host–guest interactions. The interaction pattern and binding thermodynamics show a significant dependence on the modelling parameters. The refitted system-specific parameter set improves the consistency of the modelling results and the experimental reference significantly. Finally, combining the previous charge-scheme comparison and the current force-field refitting, we provide general guidelines for the theoretical modelling of host–guest binding.
Collapse
|
38
|
Admane N, Kothandan R, Syed S, Biswas S. A quinoline alkaloid potentially modulates the amyloidogenic structural transitions of the biofilm scaffolding small basic protein. J Biomol Struct Dyn 2023; 41:1366-1377. [PMID: 34963419 DOI: 10.1080/07391102.2021.2020165] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Bacterial biofilm formation by communities of opportunistic bacterial pathogens like Staphylococcus epidermidis is regarded as the primary virulence mechanism facilitating the spread of detrimental nosocomial and implant-associated infections. An 18-kDa small basic protein (Sbp) and its amyloid fibrils account for strengthening the biofilm architecture and scaffolding the S. epidermidis biofilm matrix. Our study reports systematic analysis of the amyloidogenic structural transitions of Sbp and predicts the amyloid core of the protein which may trigger misfolding and aggregation. Herein, we report the novel amyloid inhibitory potential of Camptothecin, a quinoline alkaloid which binds stably to Sbp monomers and redirects the formation of unstructured regions further destabilizing the protein. Molecular dynamics simulations reveal that Camptothecin averts β-sheet transitions, interrupts with electrostatic interactions and disrupts the intermolecular hydrophobic associations between the exposed hydrophobic amyloidogenic regions of Sbp. Collectively, our study puts forward the first report detailing the heteromolecular associations and amyloid modulatory effects of Camptothecin which may serve as a structural scaffold for the tailored designing of novel drugs targeting the S. epidermidis biofilm matrix.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nikita Admane
- ViStA Lab, Department of Biological Sciences, BITS, Goa, India
| | - Ram Kothandan
- Bioinformatics Laboratory, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, India
| | - Sowfia Syed
- Bioinformatics Laboratory, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, India
| | - Sumit Biswas
- ViStA Lab, Department of Biological Sciences, BITS, Goa, India
| |
Collapse
|
39
|
Natarajan A, Rangan K, Vadrevu R. Self-assembly of a peptide sequence, EKKE, composed of exclusively charged amino acids: Role of charge in morphology and lead binding. J Pept Sci 2023; 29:e3451. [PMID: 36098076 DOI: 10.1002/psc.3451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/14/2022] [Accepted: 09/08/2022] [Indexed: 01/12/2023]
Abstract
The self-assembly of peptides is influenced by their amino acid sequence and other factors including pH, charge, temperature, and solvent. Herein, we explore whether a four-residue sequence, EKKE, consisting of exclusively charged amino acids shows the propensity to form self-assembled ordered nanostructures and whether the overall charge plays any role in morphological and functional properties. From a combination of experimental data provided by Thioflavin T fluorescence, Congo red absorbance, circular dichroism spectroscopy, dynamic light scattering, field emission-scanning electron microscopy, atomic force microscopy, and confocal microscopy, it is clear that the all-polar peptide and charged EKKE sequence shows a pH-dependent tendency to form amyloid-like structures, and the self-assembled entities under acidic, basic and neutral conditions exhibit morphological variation. Additionally, the ability of the self-assembled amyloid nanostructures to bind to the toxic metal, lead (Pb2+ ), was demonstrated from the analysis of the ultraviolet absorbance and X-ray photoelectron spectroscopy data. The modulation at the sequence level for the amyloid-forming EKKE scaffold can further extend its potential role not only in the remediation of other toxic metals but also towards biomedical applications.
Collapse
Affiliation(s)
- Aishwarya Natarajan
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| | - Krishnan Rangan
- Department of Chemistry, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| | - Ramakrishna Vadrevu
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| |
Collapse
|
40
|
Thursch LJ, Lima TA, O'Neill N, Ferreira FF, Schweitzer-Stenner R, Alvarez NJ. Influence of central sidechain on self-assembly of glycine-x-glycine peptides. SOFT MATTER 2023; 19:394-409. [PMID: 36454226 DOI: 10.1039/d2sm01082h] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Low molecular weight gelators (LMWGs) are the subject of intense research for a range of biomedical and engineering applications. Peptides are a special class of LMWG, which offer infinite sequence possibilities and, therefore, engineered properties. This work examines the propensity of the GxG peptide family, where x denotes a guest residue, to self-assemble into fibril networks via changes in pH and ethanol concentration. These triggers for gelation are motivated by recent work on GHG and GAG, which unexpectedly self-assemble into centimeter long fibril networks with unique rheological properties. The propensity of GxG peptides to self-assemble, and the physical and chemical properties of the self-assembled structures are characterized by microscopy, spectroscopy, rheology, and X-ray diffraction. Interestingly, we show that the number, length, size, and morphology of the crystalline self-assembled aggregates depend significantly on the x-residue chemistry and the solution conditions, i.e. pH, temperature, peptide concentration, etc. The different x-residues allow us to probe the importance of different peptide interactions, e.g. π-π stacking, hydrogen bonding, and hydrophobicity, on the formation of fibrils. We conclude that fibril formation requires π-π stacking interactions in pure water, while hydrogen bonding can form fibrils in the presence of ethanol-water solutions. These results validate and support theoretical arguments on the propensity for self-assembly and leads to a better understanding of the relationship between peptide chemistry and fibril self-assembly. Overall, GxG peptides constitute a unique family of peptides, whose characterization will aid in advancing our understanding of self-assembly driving forces for fibril formation in peptide systems.
Collapse
Affiliation(s)
- Lavenia J Thursch
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
| | - Thamires A Lima
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
| | - Nichole O'Neill
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, USA.
| | - Fabio F Ferreira
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo, Brazil
| | | | - Nicolas J Alvarez
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Rosetti B, Marchesan S. Peptide Inhibitors of Insulin Fibrillation: Current and Future Challenges. Int J Mol Sci 2023; 24:1306. [PMID: 36674821 PMCID: PMC9863703 DOI: 10.3390/ijms24021306] [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: 12/09/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Amyloidoses include a large variety of local and systemic diseases that share the common feature of protein unfolding or refolding into amyloid fibrils. The most studied amyloids are those directly involved in neurodegenerative diseases, while others, such as those formed by insulin, are surprisingly far less studied. Insulin is a very important polypeptide that plays a variety of biological roles and, first and foremost, is at the basis of the therapy of diabetic patients. It is well-known that it can form fibrils at the site of injection, leading to inflammation and immune response, in addition to other side effects. In this concise review, we analyze the current knowledge on insulin fibrillation, with a focus on the development of peptide-based inhibitors, which are promising candidates for their biocompatibility but still pose challenges to their effective use in therapy.
Collapse
Affiliation(s)
| | - Silvia Marchesan
- Chemical and Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
42
|
Al-Hilaly YK, Marshall KE, Lutter L, Biasetti L, Mengham K, Harrington CR, Xue WF, Wischik CM, Serpell LC. An Additive-Free Model for Tau Self-Assembly. Methods Mol Biol 2023; 2551:163-188. [PMID: 36310203 DOI: 10.1007/978-1-0716-2597-2_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Tau is a natively unfolded protein that contributes to the stability of microtubules. Under pathological conditions such as Alzheimer's disease (AD), tau protein misfolds and self-assembles to form paired helical filaments (PHFs) and straight filaments (SFs). Full-length tau protein assembles poorly and its self-assembly is enhanced with polyanions such as heparin and RNA in vitro, but a role for heparin or other polyanions in vivo remains unclear. Recently, a truncated form of tau (297-391) has been shown to self-assemble in the absence of additives which provides an alternative in vitro PHF model system. Here we describe methods to prepare in vitro PHFs and SFs from tau (297-391) named dGAE. We also discuss the range of biophysical/biochemical techniques used to monitor tau filament assembly and structure.
Collapse
Affiliation(s)
- Youssra K Al-Hilaly
- Chemistry Department, College of Science, Mustansiriyah University, Baghdad, Iraq.
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK.
| | - Karen E Marshall
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| | - Liisa Lutter
- School of Biosciences, University of Kent, Canterbury, UK
- Institute of Genomics and Proteomics, University of California, Los Angeles, CA, USA
| | - Luca Biasetti
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| | - Kurtis Mengham
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| | - Charles R Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- TauRx Therapeutics Ltd., Aberdeen, UK
| | - Wei-Feng Xue
- School of Biosciences, University of Kent, Canterbury, UK
| | - Claude M Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- TauRx Therapeutics Ltd., Aberdeen, UK
| | - Louise C Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| |
Collapse
|
43
|
Mohammed AS, Uversky VN. Intrinsic Disorder as a Natural Preservative: High Levels of Intrinsic Disorder in Proteins Found in the 2600-Year-Old Human Brain. BIOLOGY 2022; 11:1704. [PMID: 36552214 PMCID: PMC9775155 DOI: 10.3390/biology11121704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
Proteomic analysis revealed the preservation of many proteins in the Heslington brain (which is at least 2600-year-old brain tissue uncovered within the skull excavated in 2008 from a pit in Heslington, Yorkshire, England). Five of these proteins-"main proteins": heavy, medium, and light neurofilament proteins (NFH, NFM, and NFL), glial fibrillary acidic protein (GFAP), and myelin basic (MBP) protein-are engaged in the formation of non-amyloid protein aggregates, such as intermediate filaments and myelin sheath. We used a wide spectrum of bioinformatics tools to evaluate the prevalence of functional disorder in several related sets of proteins, such as the main proteins and their 44 interactors, all other proteins identified in the Heslington brain, as well as the entire human proteome (20,317 manually curated proteins), and 10,611 brain proteins. These analyses revealed that all five main proteins, half of their interactors and almost one third of the Heslington brain proteins are expected to be mostly disordered. Furthermore, most of the remaining Heslington brain proteins are expected to contain sizable levels of disorder. This is contrary to the expected substantial (if not complete) elimination of the disordered proteins from the Heslington brain. Therefore, it seems that the intrinsic disorder of NFH, NFM, NFL, GFAP, and MBP, their interactors, and many other proteins might play a crucial role in preserving the Heslington brain by forming tightly folded brain protein aggregates, in which different parts are glued together via the disorder-to-order transitions.
Collapse
Affiliation(s)
- Aaron S. Mohammed
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd. MDC07, Tampa, FL 33612, USA
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd. MDC07, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
44
|
Alraawi Z, Banerjee N, Mohanty S, Kumar TKS. Amyloidogenesis: What Do We Know So Far? Int J Mol Sci 2022; 23:ijms232213970. [PMID: 36430450 PMCID: PMC9695042 DOI: 10.3390/ijms232213970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
The study of protein aggregation, and amyloidosis in particular, has gained considerable interest in recent times. Several neurodegenerative diseases, such as Alzheimer's (AD) and Parkinson's (PD) show a characteristic buildup of proteinaceous aggregates in several organs, especially the brain. Despite the enormous upsurge in research articles in this arena, it would not be incorrect to say that we still lack a crystal-clear idea surrounding these notorious aggregates. In this review, we attempt to present a holistic picture on protein aggregation and amyloids in particular. Using a chronological order of discoveries, we present the case of amyloids right from the onset of their discovery, various biophysical techniques, including analysis of the structure, the mechanisms and kinetics of the formation of amyloids. We have discussed important questions on whether aggregation and amyloidosis are restricted to a subset of specific proteins or more broadly influenced by the biophysiochemical and cellular environment. The therapeutic strategies and the significant failure rate of drugs in clinical trials pertaining to these neurodegenerative diseases have been also discussed at length. At a time when the COVID-19 pandemic has hit the globe hard, the review also discusses the plausibility of the far-reaching consequences posed by the virus, such as triggering early onset of amyloidosis. Finally, the application(s) of amyloids as useful biomaterials has also been discussed briefly in this review.
Collapse
Affiliation(s)
- Zeina Alraawi
- Department of Chemistry and Biochemistry, Fulbright College of Art and Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Nayan Banerjee
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Srujana Mohanty
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata 741246, India
| | | |
Collapse
|
45
|
Jayawardena BM, Peacey L, Gamsjaeger R, Jones CE. Essential Role of Histidine for Rapid Copper(II)-Mediated Disassembly of Neurokinin B Amyloid. Biomolecules 2022; 12:biom12111585. [PMID: 36358935 PMCID: PMC9687585 DOI: 10.3390/biom12111585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 12/02/2022] Open
Abstract
Neurokinin B is a tachykinin peptide involved in a diverse range of neuronal functions. It rapidly forms an amyloid, which is considered physiologically important for efficient packing into dense core secretory vesicles within hypothalamic neurons. Disassembly of the amyloid is thought to require the presence of copper ions, which interact with histidine at the third position in the peptide sequence. However, it is unclear how the histidine is involved in the amyloid structure and why copper coordination can trigger disassembly. In this work, we demonstrate that histidine contributes to the amyloid structure via π-stacking interactions with nearby phenylalanine residues. The ability of neurokinin B to form an amyloid is dependent on any aromatic residue at the third position in the sequence; however, only the presence of histidine leads to both amyloid formation and rapid copper-induced disassembly.
Collapse
|
46
|
Liu X, Zheng L, Qin C, Zhang JZH, Sun Z. Comprehensive evaluation of end-point free energy techniques in carboxylated-pillar[6]arene host-guest binding: I. Standard procedure. J Comput Aided Mol Des 2022; 36:735-752. [PMID: 36136209 DOI: 10.1007/s10822-022-00475-0] [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: 07/06/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
Despite the massive application of end-point free energy methods in protein-ligand and protein-protein interactions, computational understandings about their performance in relatively simple and prototypical host-guest systems are limited. In this work, we present a comprehensive benchmark calculation with standard end-point free energy techniques in a recent host-guest dataset containing 13 host-guest pairs involving the carboxylated-pillar[6]arene host. We first assess the charge schemes for solutes by comparing the charge-produced electrostatics with many ab initio references, in order to obtain a preliminary albeit detailed view of the charge quality. Then, we focus on four modelling details of end-point free energy calculations, including the docking procedure for the generation of initial condition, the charge scheme for host and guest molecules, the water model used in explicit-solvent sampling, and the end-point methods for free energy estimation. The binding thermodynamics obtained with different modelling schemes are compared with experimental references, and some practical guidelines on maximizing the performance of end-point methods in practical host-guest systems are summarized. Further, we compare our simulation outcome with predictions in the grand challenge and discuss further developments to improve the prediction quality of end-point free energy methods. Overall, unlike the widely acknowledged applicability in protein-ligand binding, the standard end-point calculations cannot produce useful outcomes in host-guest binding and thus are not recommended unless alterations are performed.
Collapse
Affiliation(s)
- Xiao Liu
- School of Mathematics, Physics and Statistics, Shanghai University of Engineering Science, Shanghai, 201620, China.
| | - Lei Zheng
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, 200062, China
| | - Chu Qin
- School of Mathematics, Physics and Statistics, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - John Z H Zhang
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, 200062, China.,School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China.,Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.,Department of Chemistry, New York University, New York, NY, 10003, USA
| | - Zhaoxi Sun
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
47
|
Weiffert T, Meisl G, Curk S, Cukalevski R, Šarić A, Knowles TPJ, Linse S. Influence of denaturants on amyloid β42 aggregation kinetics. Front Neurosci 2022; 16:943355. [PMID: 36203800 PMCID: PMC9531139 DOI: 10.3389/fnins.2022.943355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
Amyloid formation is linked to devastating neurodegenerative diseases, motivating detailed studies of the mechanisms of amyloid formation. For Aβ, the peptide associated with Alzheimer's disease, the mechanism and rate of aggregation have been established for a range of variants and conditions in vitro and in bodily fluids. A key outstanding question is how the relative stabilities of monomers, fibrils and intermediates affect each step in the fibril formation process. By monitoring the kinetics of aggregation of Aβ42, in the presence of urea or guanidinium hydrochloride (GuHCl), we here determine the rates of the underlying microscopic steps and establish the importance of changes in relative stability induced by the presence of denaturant for each individual step. Denaturants shift the equilibrium towards the unfolded state of each species. We find that a non-ionic denaturant, urea, reduces the overall aggregation rate, and that the effect on nucleation is stronger than the effect on elongation. Urea reduces the rate of secondary nucleation by decreasing the coverage of fibril surfaces and the rate of nucleus formation. It also reduces the rate of primary nucleation, increasing its reaction order. The ionic denaturant, GuHCl, accelerates the aggregation at low denaturant concentrations and decelerates the aggregation at high denaturant concentrations. Below approximately 0.25 M GuHCl, the screening of repulsive electrostatic interactions between peptides by the charged denaturant dominates, leading to an increased aggregation rate. At higher GuHCl concentrations, the electrostatic repulsion is completely screened, and the denaturing effect dominates. The results illustrate how the differential effects of denaturants on stability of monomer, oligomer and fibril translate to differential effects on microscopic steps, with the rate of nucleation being most strongly reduced.
Collapse
Affiliation(s)
- Tanja Weiffert
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Georg Meisl
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Samo Curk
- Department of Physics and Astronomy, University College London, London, United Kingdom
| | - Risto Cukalevski
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Anđela Šarić
- Department of Physics and Astronomy, University College London, London, United Kingdom
| | - Tuomas P. J. Knowles
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - Sara Linse
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| |
Collapse
|
48
|
Buell AK. Stability matters, too - the thermodynamics of amyloid fibril formation. Chem Sci 2022; 13:10177-10192. [PMID: 36277637 PMCID: PMC9473512 DOI: 10.1039/d1sc06782f] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/30/2022] [Indexed: 12/26/2022] Open
Abstract
Amyloid fibrils are supramolecular homopolymers of proteins that play important roles in biological functions and disease. These objects have received an exponential increase in attention during the last few decades, due to their role in the aetiology of a range of severe disorders, most notably some of a neurodegenerative nature. While an overwhelming number of experimental studies exist that investigate how, and how fast, amyloid fibrils form and how their formation can be inhibited, a much more limited body of experimental work attempts to answer the question as to why these types of structures form (i.e. the thermodynamic driving force) and how stable they actually are. In this review, I attempt to give an overview of the types of experiments that have been performed to-date to answer these questions, and to summarise our current understanding of amyloid thermodynamics.
Collapse
Affiliation(s)
- Alexander K Buell
- Technical University of Denmark, Department of Biotechnology and Biomedicine Søltofts Plads, Building 227 2800 Kgs. Lyngby Denmark
| |
Collapse
|
49
|
Mocanu CS, Darie-Ion L, Petre BA, Gradinaru VR, Drochioiu G. A computational study of metal ions interaction with amyloid-β 1-42 peptide structure in hyperpyrexia: Implications for Alzheimer disease. JOURNAL OF KING SAUD UNIVERSITY. SCIENCE 2022; 34:102184. [PMID: 35783243 PMCID: PMC9238029 DOI: 10.1016/j.jksus.2022.102184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 05/28/2023]
Abstract
Given the current context of the SARS-CoV-19 pandemic, among the interfering risky factors with the Aβ peptide aggregation in the brains of Alzheimer's disease (AD) patients can be hyperpyrexia and increased intracranial pressure (ICP). According to our hypothesis on the relationship between hyperpyrexia and cognitive decline in AD, two models of Aβ peptides were used in this study: the structure of AD amyloid beta-peptide and near-atomic resolution fibril structures of the Aβ peptide. Therefore, the binding templates were constructed for Aβ peptide regions able to bind 9 different metal ions. The fragment transformation method was used for the structural comparison between Aβ chains. Molecular dynamics simulation (MDS) was applied using the Nose-Poincare-Anderson equation to generate a theoretically correct NPT (isothermal-isobaric ensemble). The smallest dissimilarities were observed in the case of Cu+ binding potential followed by Co2+, both with similar variation. Structural changes have also occurred as a result of the dynamic simulation. All these changes suggest an aggravating factor in both hyperpyretic and AD conditions. Our findings suggest that elevated temperature and increased intracranial pressure rise the effect of peptide aggregation, by converting α-helix motif to β-sheet and random coil conformation, which are related to the formation of senile plaques in AD brains.
Collapse
Affiliation(s)
- Cosmin Stefan Mocanu
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| | - Laura Darie-Ion
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| | - Brindusa Alina Petre
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
- Center for Fundamental Research and Experimental Development in Translation Medicine, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot Str., 700483 Iasi, Romania
| | | | - Gabi Drochioiu
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| |
Collapse
|
50
|
Takekiyo T, Yamada N, Amo T, Asano A, Yoshimura Y. Triiodide ion-induced inhibition of amyloid aggregate formation: A case study of α-synuclein. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|