1
|
Sehra N, Parmar R, Jain R. Peptide-based amyloid-beta aggregation inhibitors. RSC Med Chem 2024:d4md00729h. [PMID: 39882170 PMCID: PMC11773382 DOI: 10.1039/d4md00729h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/28/2024] [Indexed: 01/31/2025] Open
Abstract
Aberrant protein misfolding and accumulation is considered to be a major pathological pillar of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Aggregation of amyloid-β (Aβ) peptide leads to the formation of toxic amyloid fibrils and is associated with cognitive dysfunction and memory loss in Alzheimer's disease (AD). Designing molecules that inhibit amyloid aggregation seems to be a rational approach to AD drug development. Over the years, researchers have utilized a variety of therapeutic strategies targeting different pathways, extensively studying peptide-based approaches to understand AD pathology and demonstrate their efficacy against Aβ aggregation. This review highlights rationally designed peptide/mimetics, including structure-based peptides, metal-peptide chelators, stapled peptides, and peptide-based nanomaterials as potential amyloid inhibitors.
Collapse
Affiliation(s)
- Naina Sehra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Sector 67, S. A. S. Nagar Punjab 160062 India
| | - Rajesh Parmar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Sector 67, S. A. S. Nagar Punjab 160062 India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Sector 67, S. A. S. Nagar Punjab 160062 India
| |
Collapse
|
2
|
Sehra N, Parmar R, Maurya IK, Kumar V, Tikoo K, Jain R. Synthesis and mechanistic study of Aβ 42 C-terminus domain derived tetrapeptides that inhibit Alzheimer's Aβ-aggregation-induced neurotoxicity. Bioorg Med Chem Lett 2024; 112:129929. [PMID: 39151661 DOI: 10.1016/j.bmcl.2024.129929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Amyloid plaque formation in the brain is mainly responsible for the onset of Alzheimer's disease (AD). Structure-based peptides have gained importance in recent years, and rational design of the peptide sequences for the prevention of Aβ-aggregation and related toxicity is imperative. In this study, we investigate the structural modification of tetrapeptides derived from the hydrophobic C-terminal region of Aβ42 "VVIA-NH2" and its retro-sequence "AIVV-NH2." A preliminary screening of synthesized peptides through an MTT cell viability assay followed by a ThT fluorescence assay revealed a peptide 13 (Ala-Ile-Aib-Val-NH2) that showed protection against Aβ-aggregation and associated neurotoxicity. The presence of the α-helix inducer "Aib" in peptide 13 manifested the conformational transition from cross-β-sheets to α-helical content in Aβ42. The absence of fibrils in electron microscopic analysis suggested the inhibitory potential of peptide 13. The HRMS, DLS, and ANS studies further confirmed the inhibitory activity of 13, and no cytotoxicity was observed. The structure-based peptide described herein is a promising amyloid-β inhibitor and provides a new lead for the development of AD therapeutics.
Collapse
Affiliation(s)
- Naina Sehra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Rajesh Parmar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Indresh K Maurya
- Center of Infectious Disease, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Vinod Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
3
|
Picone P, Sanfilippo T, Vasto S, Baldassano S, Guggino R, Nuzzo D, Bulone D, San Biagio PL, Muscolino E, Monastero R, Dispenza C, Giacomazza D. From Small Peptides to Large Proteins against Alzheimer’sDisease. Biomolecules 2022; 12:biom12101344. [PMID: 36291553 PMCID: PMC9599460 DOI: 10.3390/biom12101344] [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: 07/14/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder in the elderly. The two cardinal neuropathological hallmarks of AD are the senile plaques, which are extracellular deposits mainly constituted by beta-amyloids, and neurofibrillary tangles formed by abnormally phosphorylated Tau (p-Tau) located in the cytoplasm of neurons. Although the research has made relevant progress in the management of the disease, the treatment is still lacking. Only symptomatic medications exist for the disease, and, in the meantime, laboratories worldwide are investigating disease-modifying treatments for AD. In the present review, results centered on the use of peptides of different sizes involved in AD are presented.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto per la Ricerca e l’Innovazione Biomedica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Tiziana Sanfilippo
- Ambulatorio di Nutrizione Clinica ASP Palermo, Via G. Cusmano 24, 90141 Palermo, Italy
- Anestesia e Rianimazione, Presidio Ospedaliero “S. Cimino”, 90141 Termini Imerese, Italy
| | - Sonya Vasto
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
- Istituti Euro-Mediterranei di Scienza e Tecnologia (IEMEST), Via M. Miraglia 20, 90139 Palermo, Italy
| | - Sara Baldassano
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Rossella Guggino
- Ambulatorio di Nutrizione Clinica ASP Palermo, Via G. Cusmano 24, 90141 Palermo, Italy
- Anestesia e Rianimazione, Presidio Ospedaliero “S. Cimino”, 90141 Termini Imerese, Italy
| | - Domenico Nuzzo
- Istituto per la Ricerca e l’Innovazione Biomedica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
- Correspondence: (D.N.); (D.G.)
| | - Donatella Bulone
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Pier Luigi San Biagio
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Emanuela Muscolino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze, Bldg 6, 90128 Palermo, Italy
| | - Roberto Monastero
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Clelia Dispenza
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze, Bldg 6, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Correspondence: (D.N.); (D.G.)
| |
Collapse
|
4
|
Quercetin binding accelerates prion fibrillation into proteinase sensitive and loosely structured amyloids. Biomed Pharmacother 2022; 151:113177. [PMID: 35676783 DOI: 10.1016/j.biopha.2022.113177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 11/23/2022] Open
Abstract
Amyloidoses are caused by the deposition of amyloid fibrils ascribed to protein misfolding. In this study, we examined the antiamyloidogenic and antioxidative activities of quercetin, a plant flavonol from the flavonoid group of polyphenols, on mouse prion protein (moPrP) with biophysical approaches. As the results show, quercetin binds to the C-terminal region of moPrP, and quercetin binding does not affect the structure of moPrP. However, quercetin binding accelerates moPrP fibrillation and changes the structure of moPrP fibrils. Unlike typical prion fibrils, quercetin-bound fibrils are sensitive to proteinase K and are loosely structured. Moreover, due to high antioxidant activity of flavonoid, quercetin-bound fibrils lack imbalance of free radicals and, therefore, they are nontoxic towards neuroblastoma cells. The quercetin shows its uniqueness from typical antiamyloidogenic drugs which either suppress the development of amyloid or eliminate formed amyloids. Quercetin binding converts moPrP into protease-sensitive and non-cytotoxic fibrils. This work provides a powerful resolution in the advancement of antiamyloidogenic treatment.
Collapse
|
5
|
Kumari M, Sharma S, Deep S. Tetrabutylammonium based ionic liquids (ILs) inhibit the amyloid aggregation of superoxide dismutase 1 (SOD1). J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.118761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
6
|
Santoro A, Grimaldi M, Buonocore M, Stillitano I, Gloria A, Santin M, Bobba F, Sublimi Saponetti M, Ciaglia E, D'Ursi AM. New Aβ(1-42) ligands from anti-amyloid antibodies: Design, synthesis, and structural interaction. Eur J Med Chem 2022; 237:114400. [PMID: 35489223 DOI: 10.1016/j.ejmech.2022.114400] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/17/2022] [Accepted: 04/15/2022] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD), is the most common neurodegenerative disorder of the aging population resulting in progressive cognitive and functional decline. Accumulation of amyloid plaques around neuronal cells is considered a critical pathogenetic event and, in most cases, a hallmark of the pathology. In the attempt to identify anti-AD drug candidates, hundreds of molecules targeting Aβ peptides have been screened. Peptide molecules have been widely explored, appreciating chemical stability, biocompatibility, and low production cost. More recently, many anti-Aβ(1-42) monoclonal antibodies have been developed, given the excellent potential of immunotherapy for treating or preventing AD. Antibodies are versatile ligands that bind a large variety of molecules with high affinity and specificity; however, their extensive therapeutic application is complex and requires huge economic investments. Novel approaches to identify alternative antibody formats are considered with great interest. In this context, taking advantage of the favorable peptide properties and the availability of Aβ-antibodies structural data, we followed an innovative research approach to identify short peptide sequences on the model of the binding sites of Aβ(1-42)/antibodies. WAibH and SYSTPGK were designed as mimics of solanezumab and aducanumab, respectively. Circular dichroism and nuclear magnetic resonance analysis reveal that the antibody-derived peptides interact with Aβ(1-42) in the soluble monomeric form. Moreover, AFM microscopy imaging shows that WAibH and SYSTPGK are capable of controlling the Aβ(1-42) aggregation. The strategy to identify WAibH and SYSTPGK is innovative and can be widely applied for new anti-Aβ antibody mimicking peptides.
Collapse
Affiliation(s)
- Angelo Santoro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Michela Buonocore
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Ilaria Stillitano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, V.le J.F. Kennedy 54 - Pad. 20, Mostra d'Oltremare, 80125, Naples, Italy
| | - Matteo Santin
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, BN2 4GJ, UK
| | - Fabrizio Bobba
- Department of Physics, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Matilde Sublimi Saponetti
- Department of Physics, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry Scuola Medica Salernitana, University of Salerno, Via Salvatore Allende, 84081, Baronissi, Salerno, Italy
| | - Anna Maria D'Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132 - 84084, Fisciano, Salerno, Italy.
| |
Collapse
|
7
|
de Oliveira OV, Gonçalves ADS, Almeida NECD. Insights into β-amyloid transition prevention by cucurbit[7]uril from molecular modeling. J Biomol Struct Dyn 2022; 40:9602-9612. [PMID: 34042019 DOI: 10.1080/07391102.2021.1932600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this study, comparable molecular dynamic (MD) simulations of 1.2 microseconds were performed to clarify the prevention of the β-amyloid peptide (Aβ1-42) aggregation by cucurbit[7]uril (CB[7]). The accumulation of this peptide in the brain is one of the most harmful in Alzheimer's disease. The inhibition mechanism of Aβ1-42 aggregation by different molecules is attributed to preventing of Aβ1-42 conformational transition from α-helix to the β-sheet structure. However, our structural analysis shows that the pure water and aqueous solution of the CB[7] denature the native Aβ1-42 α-helix structure forming different compactness and unfolded conformations, not in β-sheet form. On the other hand, in the three CB[7]@Aβ1-42 complexes, it was observed the encapsulation of N-terminal (Asp1), Lys16, and Val36 by CB[7] along the MD trajectory, and not with aromatic residues as suggested by the literature. Only in one CB[7]@Aβ1-42 complex was observed stable Asp23-Lys28 salt bridge with an average distance of 0.36 nm. All CB[7]@Aβ1-42 complexes are very stable with binding free energy lowest than ∼-50 kcal/mol between the CB[7] and Aβ1-42 monomer from MM/PBSA calculation. Therefore, herein we show that the mechanism of the prevention of elongation protofibril by CB[7] is due to the disruption of the Asp23-Lys28 salt bridge and steric effects of CB[7]@Aβ1-42 complex with the fibril lattice, and not due to the transition from α-helix to β-sheet following the dock-lock mechanism.Communicated by Ramaswamy H. Sarma.
Collapse
|
8
|
Sağlık BN, Osmaniye D, Acar Çevik U, Levent S, Kaya Çavuşoğlu B, Özkay Y, Kaplancıklı ZA. Design, Synthesis, and Structure-Activity Relationships of Thiazole Analogs as Anticholinesterase Agents for Alzheimer's Disease. Molecules 2020; 25:E4312. [PMID: 32962239 PMCID: PMC7570694 DOI: 10.3390/molecules25184312] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/10/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022] Open
Abstract
Dementia is a neurological condition commonly correlated with Alzheimer's disease (AD), and it is seen with many other central nervous system (CNS) disorders. The restricted number of medications is not appropriate to offer enough relief to enhance the quality of life of patients suffering from this symptom; thus, all therapeutic choices should be carefully assessed. In this study, new thiazolylhydrazone derivatives (2a-2l) were designed and synthesized based on the cholinergic hypothesis. Their chemical structures were confirmed by 1H NMR, 13C NMR, and HRMS spectrometric techniques. The ADME (absorption, distribution, metabolism, elimination) parameters of the synthesized compounds were predicted by using QikProp 4.8 software. It was concluded that all compounds presented satisfactory drug-like characteristics. Furthermore, their inhibitory activities against acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) in vitro were also tested by modified the Ellman spectrophotometric method. According to the results, all compounds showed a weak inhibitory effect on BChE. On the other hand, most of the compounds (2a, 2b, 2d, 2e, 2g, 2i, and 2j) had a certain AChE inhibitory activity, and the IC50 values of them were calculated as 0.063 ± 0.003, 0.056 ± 0.002, 0.147 ± 0.006, 0.040 ± 0.001, 0.031 ± 0.001, 0.028 ± 0.001, and 0.138 ± 0.005 µM, respectively. Among these derivatives, compound 2i was found to be the most active agent in the series with an IC50 value of 0.028 ± 0.001 µM, which indicated an inhibition profile at a similar rate as the reference drug, donepezil. The potential binding modes of compounds 2a, 2b, 2e, 2g, and 2i with AChE were investigated and compared with each other by the molecular docking studies. The results showed that these compounds were strongly bound up with the AChE enzyme active site with the optimal conformations.
Collapse
Affiliation(s)
- Begüm Nurpelin Sağlık
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; (B.N.S.); (D.O.); (U.A.Ç.); (S.L.); (Y.Ö.)
- Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| | - Derya Osmaniye
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; (B.N.S.); (D.O.); (U.A.Ç.); (S.L.); (Y.Ö.)
- Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| | - Ulviye Acar Çevik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; (B.N.S.); (D.O.); (U.A.Ç.); (S.L.); (Y.Ö.)
- Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| | - Serkan Levent
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; (B.N.S.); (D.O.); (U.A.Ç.); (S.L.); (Y.Ö.)
- Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| | - Betül Kaya Çavuşoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Zonguldak Bülent Ecevit University, 67600 Zonguldak, Turkey;
| | - Yusuf Özkay
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; (B.N.S.); (D.O.); (U.A.Ç.); (S.L.); (Y.Ö.)
- Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| | - Zafer Asım Kaplancıklı
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; (B.N.S.); (D.O.); (U.A.Ç.); (S.L.); (Y.Ö.)
| |
Collapse
|
9
|
Bhatia NK, Modi P, Sharma S, Deep S. Quercetin and Baicalein Act as Potent Antiamyloidogenic and Fibril Destabilizing Agents for SOD1 Fibrils. ACS Chem Neurosci 2020; 11:1129-1138. [PMID: 32208672 DOI: 10.1021/acschemneuro.9b00677] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that has been associated with the deposition of aggregates of superoxide dismutase 1 (SOD1). Effective therapeutics against SOD1 fibrillation is still an area of active research. Herein, we demonstrate the potential of two naturally occurring flavonoids (quercetin and baicalein) to inhibit fibrillation of wild-type SOD1 with the aid of a series of biophysical techniques. Our seeding experiments reveal that both of these flavonoids significantly affect the fibril elongation. Interestingly, our ThT binding assay, TEM, and SDS-PAGE experiments suggest that these flavonoids also disintegrate the fibrils into shorter fragments but do not completely depolymerize them into monomers. Binding parameters obtained from the analysis of UV-vis spectra suggest that these flavonoids bind moderately to native SOD1 dimer and have different binding sites. Docking of these flavonoids with a non-native monomer, non-native trimer, and oligomer derived from the 11-residue segment of SOD1 indicates that both quercetin and baicalein can bind to these species and thus can arrest the elongation of fibrils by blocking the fibrillar core regions on the intermediate species formed during aggregation of SOD1. MTT assay data revealed that both the flavonoids reduced the cytotoxicity of SOD1 fibrils. Experimental data also show the antiamyloidogenic potential of both flavonoids against A4V SOD1 mutant fibrillation. Thus, our findings may provide a direction for designing effective therapeutic agents against ALS which can act as promising antiamyloidogenic and fibril destabilizing agents.
Collapse
Affiliation(s)
- Nidhi K. Bhatia
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Priya Modi
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Shilpa Sharma
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
10
|
Lazzaro S, Ogrinc N, Lamont L, Vecchio G, Pappalardo G, Heeren RMA. Ion mobility spectrometry combined with multivariate statistical analysis: revealing the effects of a drug candidate for Alzheimer's disease on Aβ1-40 peptide early assembly. Anal Bioanal Chem 2019; 411:6353-6363. [PMID: 31407050 PMCID: PMC6718366 DOI: 10.1007/s00216-019-02030-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/20/2022]
Abstract
Inhibition of the initial stages of amyloid-β peptide self-assembly is a key approach in drug development for Alzheimer's disease, in which soluble and highly neurotoxic low molecular weight oligomers are produced and aggregate in the brain over time. Here we report a high-throughput method based on ion mobility mass spectrometry and multivariate statistical analysis to rapidly select statistically significant early-stage species of amyloid-β1-40 whose formation is inhibited by a candidate theranostic agent. Using this method, we have confirmed the inhibition of a Zn-porphyrin-peptide conjugate in the early self-assembly of Aβ40 peptide. The MS/MS fragmentation patterns of the species detected in the samples containing the Zn-porphyrin-peptide conjugate suggested a porphyrin-catalyzed oxidation at Met-35(O) of Aβ40. We introduce ion mobility MS combined with multivariate statistics as a systematic approach to perform data analytics in drug discovery/amyloid research that aims at the evaluation of the inhibitory effect on the Aβ early assembly in vitro models at very low concentration levels of Aβ peptides.
Collapse
Affiliation(s)
- Serena Lazzaro
- Institute of Biostructures and Bioimaging (IBB), National Research Council, Via Paolo Gaifami N.18, 95126, Catania, Italy
| | - Nina Ogrinc
- The Maastricht Multimodal Molecular Imaging institute M4I- Division of Imaging Mass Spectrometry, Maastricht University, Minderbroedersberg 4-6, 6211 LK, Maastricht, The Netherlands
| | - Lieke Lamont
- The Maastricht Multimodal Molecular Imaging institute M4I- Division of Imaging Mass Spectrometry, Maastricht University, Minderbroedersberg 4-6, 6211 LK, Maastricht, The Netherlands
| | - Graziella Vecchio
- Department of Chemical Sciences, Catania University, Viale Andrea Doria, 6, 95125, Catania, Italy
| | - Giuseppe Pappalardo
- Institute of Biostructures and Bioimaging (IBB), National Research Council, Via Paolo Gaifami N.18, 95126, Catania, Italy
| | - Ron M A Heeren
- The Maastricht Multimodal Molecular Imaging institute M4I- Division of Imaging Mass Spectrometry, Maastricht University, Minderbroedersberg 4-6, 6211 LK, Maastricht, The Netherlands.
| |
Collapse
|
11
|
Design and Synthesis of 2-Substitutedphenyl Benzo[D]Thiazole Derivatives and Their β-Amyloid Aggregation and Cholinesterase Inhibitory Activities. Pharm Chem J 2019. [DOI: 10.1007/s11094-019-02000-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
12
|
Ye Z, Wei L, Li Y, Xiao L. Efficient Modulation of β-Amyloid Peptide Fibrillation with Polymer Nanoparticles Revealed by Super-Resolution Optical Microscopy. Anal Chem 2019; 91:8582-8590. [PMID: 31148450 DOI: 10.1021/acs.analchem.9b01877] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
β-Amyloid peptide (Aβ) aggregation is the essential hallmark of neurodegenerative disorders such as Alzheimer's disease. Efficient inhibitors are highly desired for the prevention of Aβ assembly that has been considered as the primary therapeutic strategy for neurodegenerative diseases. Apart from this, visualization of the aggregates and morphology at high spatial resolution is widely considered of crucial significance on biological treatment. In this work, we have developed small-sized (with diameter of ∼4.7 nm) and positively charged fluorescent conjugated polymer nanoparticles (CPNPs) with strong inhibition effect on Aβ1-40 peptides fibrillation. Interestingly, the CPNPs also possess excellent photophysical properties, including high photon counts, robust blinking, and repetitive fluorescence switching, that are especially suitable for localization-based super-resolution imaging. Spatial resolution of ∼20 nm for these blinking CPNPs is readily achieved. According to the optical microscopic results, it was found that binding of CPNPs to the terminal of seed fibrils can effectively inhibit the fibrillation process. Owing to these attractive biological and unique photophysical properties, the small-sized CPNPs show high potential in a variety of super-resolution based biological applications.
Collapse
Affiliation(s)
- Zhongju Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin , 300071 , China
| | - Lin Wei
- Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering , Hunan Normal University , Changsha , 410081 , China
| | - Yiliang Li
- Department of Rehabilitation Medicine , The Affiliated Baoan Hospital of Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The People's Hospital of Baoan Shenzhen , Shenzhen , 510530 , China
| | - Lehui Xiao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin , 300071 , China
| |
Collapse
|
13
|
Adessi TG, Borioni JL, Pigni NB, Bastida J, Cavallaro V, Murray AP, Puiatti M, Oberti JC, Leiva S, Nicotra VE, Garcia ME. Clinanthus microstephium, an Amaryllidaceae Species with Cholinesterase Inhibitor Alkaloids: Structure-Activity Analysis of Haemanthamine Skeleton Derivatives. Chem Biodivers 2019; 16:e1800662. [PMID: 30801949 DOI: 10.1002/cbdv.201800662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/22/2019] [Indexed: 12/14/2022]
Abstract
Plants of the Amaryllidaceae family are well-known (not only) for their ornamental value but also for the alkaloids that they produce. In this report, the first phytochemical study of Clinanthus genus was carried out. The chemical composition of alkaloid fractions from Clinanthus microstephium was analyzed by GC/MS and NMR. Seven known compounds belonging to three structural types of Amaryllidaceae alkaloids were identified. An epimeric mixture of a haemanthamine-type compound (6-hydroxymaritidine) was tested as an inhibitor against acetyl- and butyrylcholinesterase enzymes (AChE and BChE, respectively), two enzymes relevant in the treatment of Alzheimer's disease, with good results. Structure-activity relationships through molecular docking studies with this alkaloid and other structurally related compounds were discussed.
Collapse
Affiliation(s)
- Tonino G Adessi
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, XUA5000, Argentina.,Instituto Multidisciplinario de Biología Vegetal, IMBIV-CONICET, Córdoba, XUA5000, Argentina
| | - José L Borioni
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, XUA5000, Argentina.,Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, M5502JMA, Argentina
| | - Natalia B Pigni
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, XUA5000, Argentina.,Instituto de Ciencia y Tecnología de Alimentos Córdoba, ICYTAC-CONICET, Córdoba, XUA5000, Argentina
| | - Jaume Bastida
- Departamento de Biología, Sanidad y Medio Ambiente, Facultad de Farmacia y Ciencias de la Alimentación, Universidad de Barcelona, 08028, Barcelona, España
| | - Valeria Cavallaro
- INQUISUR-CONICET, Departamento de Química, Universidad Nacional del Sur, Bahía Blanca, B8000CPB, Argentina
| | - Ana P Murray
- INQUISUR-CONICET, Departamento de Química, Universidad Nacional del Sur, Bahía Blanca, B8000CPB, Argentina
| | - Marcelo Puiatti
- Instituto de Investigaciones en Físico-Química de Córdoba, INFIQC-CONICET, Córdoba, XUA5000, Argentina
| | - Juan C Oberti
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, XUA5000, Argentina.,Instituto Multidisciplinario de Biología Vegetal, IMBIV-CONICET, Córdoba, XUA5000, Argentina
| | - Segundo Leiva
- Museo de Historia Natural, Universidad Privada Antenor Orrego de Trujillo, Trujillo, 13006, Perú
| | - Viviana E Nicotra
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, XUA5000, Argentina.,Instituto Multidisciplinario de Biología Vegetal, IMBIV-CONICET, Córdoba, XUA5000, Argentina
| | - Manuela E Garcia
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, XUA5000, Argentina.,Instituto Multidisciplinario de Biología Vegetal, IMBIV-CONICET, Córdoba, XUA5000, Argentina
| |
Collapse
|
14
|
Honcharenko D, Juneja A, Roshan F, Maity J, Galán-Acosta L, Biverstål H, Hjorth E, Johansson J, Fisahn A, Nilsson L, Strömberg R. Amyloid-β Peptide Targeting Peptidomimetics for Prevention of Neurotoxicity. ACS Chem Neurosci 2019; 10:1462-1477. [PMID: 30673220 DOI: 10.1021/acschemneuro.8b00485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A new generation of ligands designed to interact with the α-helix/β-strand discordant region of the amyloid-β peptide (Aβ) and to counteract its oligomerization is presented. These ligands are designed to interact with and stabilize the Aβ central helix (residues 13-26) in an α-helical conformation with increased interaction by combining properties of several first-generation ligands. The new peptide-like ligands aim at extended hydrophobic and polar contacts across the central part of the Aβ, that is, "clamping" the target. Molecular dynamics (MD) simulations of the stability of the Aβ central helix in the presence of a set of second-generation ligands were performed and revealed further stabilization of the Aβ α-helical conformation, with larger number of polar and nonpolar contacts between ligand and Aβ, compared to first-generation ligands. The synthesis of selected novel Aβ-targeting ligands was performed in solution via an active ester coupling approach or on solid-phase using an Fmoc chemistry protocol. This included incorporation of aliphatic hydrocarbon moieties, a branched triamino acid with an aliphatic hydrocarbon tail, and an amino acid with a 4'- N, N-dimethylamino-1,8-naphthalimido group in the side chain. The ability of the ligands to reduce Aβ1-42 neurotoxicity was evaluated by gamma oscillation experiments in hippocampal slice preparations. The "clamping" second-generation ligands were found to be effective antineurotoxicity agents and strongly prevented the degradation of gamma oscillations by physiological concentration of monomeric Aβ1-42 at a stoichiometric ratio.
Collapse
Affiliation(s)
- Dmytro Honcharenko
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Alok Juneja
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Firoz Roshan
- Neuronal Oscillations Laboratory, Neurogeriatrics Division, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Jyotirmoy Maity
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Lorena Galán-Acosta
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Henrik Biverstål
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14183 Stockholm, Sweden
- Department of Physical Organic Chemistry, Latvian Institute of Organic Synthesis, Riga LV-1006, Latvia
| | - Erik Hjorth
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Jan Johansson
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14183 Stockholm, Sweden
| | - André Fisahn
- Neuronal Oscillations Laboratory, Neurogeriatrics Division, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Lennart Nilsson
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Roger Strömberg
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| |
Collapse
|
15
|
Jha NN, Ranganathan S, Kumar R, Mehra S, Panigrahi R, Navalkar A, Ghosh D, Kumar A, Padinhateeri R, Maji SK. Complexation of NAC-Derived Peptide Ligands with the C-Terminus of α-Synuclein Accelerates Its Aggregation. Biochemistry 2018; 57:791-804. [DOI: 10.1021/acs.biochem.7b01090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Narendra Nath Jha
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| | | | - Rakesh Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| | - Surabhi Mehra
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| | - Rajlaxmi Panigrahi
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| | - Dhiman Ghosh
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| | | | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400 076, India
| |
Collapse
|
16
|
Ponikova S, Kubackova J, Bednarikova Z, Marek J, Demjen E, Antosova A, Musatov A, Gazova Z. Inhibition of lysozyme amyloidogenesis by phospholipids. Focus on long-chain dimyristoylphosphocholine. Biochim Biophys Acta Gen Subj 2017; 1861:2934-2943. [DOI: 10.1016/j.bbagen.2017.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/15/2017] [Accepted: 08/28/2017] [Indexed: 11/16/2022]
|
17
|
de Almeida NEC, Do TD, LaPointe NE, Tro M, Feinstein SC, Shea JE, Bowers MT. 1,2,3,4,6-penta-O-galloyl-β-D-glucopyranose Binds to the N-terminal Metal Binding Region to Inhibit Amyloid β-protein Oligomer and Fibril Formation. INTERNATIONAL JOURNAL OF MASS SPECTROMETRY 2017; 420:24-34. [PMID: 29056865 PMCID: PMC5644501 DOI: 10.1016/j.ijms.2016.09.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The early oligomerization of amyloid β-protein (Aβ) is a crucial step in the etiology of Alzheimer's disease (AD), in which soluble and highly neurotoxic oligomers are produced and accumulated inside neurons. In search of therapeutic solutions for AD treatment and prevention, potent inhibitors that remodel Aβ assembly and prevent neurotoxic oligomer formation offer a promising approach. In particular, several polyphenolic compounds have shown anti-aggregation properties and good efficacy on inhibiting oligomeric amyloid formation. 1,2,3,4,6-penta-O-galloyl-β-D-glucopyranose is a large polyphenol that has been shown to be effective at inhibiting aggregation of full-length Aβ1-40 and Aβ1-42, but has the opposite effect on the C-terminal fragment Aβ25-35. Here, we use a combination of ion mobility coupled to mass spectrometry (IMS-MS), transmission electron microscopy (TEM) and molecular dynamics (MD) simulations to elucidate the inhibitory effect of PGG on aggregation of full-length Aβ1-40 and Aβ1-42. We show that PGG interacts strongly with these two peptides, especially in their N-terminal metal binding regions, and suppresses the formation of Aβ1-40 tetramer and Aβ1-42 dodecamer. By exploring multiple facets of polyphenol-amyloid interactions, we provide a molecular basis for the opposing effects of PGG on full-length Aβ and its C-terminal fragments.
Collapse
Affiliation(s)
- Natália E. C. de Almeida
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Thanh D. Do
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Nichole E. LaPointe
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, United States
| | - Michael Tro
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Stuart C. Feinstein
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, United States
| | - Joan-Emma Shea
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Michael T. Bowers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
18
|
Zhu D, Gong G, Wang W, Du W. Disaggregation of human islet amyloid polypeptide fibril formation by ruthenium polypyridyl complexes. J Inorg Biochem 2017; 170:109-116. [DOI: 10.1016/j.jinorgbio.2017.02.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/21/2017] [Accepted: 02/09/2017] [Indexed: 11/16/2022]
|
19
|
Loureiro JA, Andrade S, Duarte A, Neves AR, Queiroz JF, Nunes C, Sevin E, Fenart L, Gosselet F, Coelho MAN, Pereira MC. Resveratrol and Grape Extract-loaded Solid Lipid Nanoparticles for the Treatment of Alzheimer's Disease. Molecules 2017; 22:E277. [PMID: 28208831 PMCID: PMC6155722 DOI: 10.3390/molecules22020277] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/26/2017] [Accepted: 02/01/2017] [Indexed: 12/11/2022] Open
Abstract
The aggregation of amyloid-β peptide (Aβ) has been linked to the formation of neuritic plaques, which are pathological hallmarks of Alzheimer's disease (AD). Various natural compounds have been suggested as therapeutics for AD. Among these compounds, resveratrol has aroused great interest due to its neuroprotective characteristics. Here, we provide evidence that grape skin and grape seed extracts increase the inhibition effect on Aβ aggregation. However, after intravenous injection, resveratrol is rapidly metabolized into both glucuronic acid and sulfate conjugations of the phenolic groups in the liver and intestinal epithelial cells (within less than 2 h), which are then eliminated. In the present study, we show that solid lipid nanoparticles (SLNs) functionalized with an antibody, the anti-transferrin receptor monoclonal antibody (OX26 mAb), can work as a possible carrier to transport the extract to target the brain. Experiments on human brain-like endothelial cells show that the cellular uptake of the OX26 SLNs is substantially more efficient than that of normal SLNs and SLNs functionalized with an unspecific antibody. As a consequence, the transcytosis ability of these different SLNs is higher when functionalized with OX-26.
Collapse
Affiliation(s)
- Joana A Loureiro
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, Porto 4500-465, Portugal.
| | - Stephanie Andrade
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, Porto 4500-465, Portugal.
| | - Ana Duarte
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, Porto 4500-465, Portugal.
| | - Ana Rute Neves
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy of the University of Porto, Porto 4050-313, Portugal.
| | - Joana Fontes Queiroz
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy of the University of Porto, Porto 4050-313, Portugal.
| | - Cláudia Nunes
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy of the University of Porto, Porto 4050-313, Portugal.
| | - Emmanuel Sevin
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens F-62300, France.
| | - Laurence Fenart
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens F-62300, France.
| | - Fabien Gosselet
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens F-62300, France.
| | - Manuel A N Coelho
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, Porto 4500-465, Portugal.
| | - Maria Carmo Pereira
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, Porto 4500-465, Portugal.
| |
Collapse
|
20
|
Profit AA, Vedad J, Desamero RZB. Peptide Conjugates of Benzene Carboxylic Acids as Agonists and Antagonists of Amylin Aggregation. Bioconjug Chem 2017; 28:666-677. [PMID: 28071890 DOI: 10.1021/acs.bioconjchem.6b00732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human islet amyloid polypeptide (hIAPP), also known as amylin, is a 37 residue peptide hormone that is stored and co-secreted with insulin. hIAPP plays a pivotal role in type 2 diabetes and is the major component of amyloid deposits found in the pancreas of patients afflicted with the disease. The self-assembly of hIAPP and the formation of amyloid is linked to the death of insulin producing β-cells. Recent findings suggest that soluble hIAPP oligomers are the cytotoxic species responsible for β-cell loss whereas amyloid fibrils themselves may indeed be innocuous. Potential avenues of therapeutic intervention include the development of compounds that prevent hIAPP self-assembly as well as those that reduce or eliminate lag time and rapidly accelerate the formation of amyloid fibrils. Both of these approaches minimize temporal exposure to soluble cytotoxic hIAPP oligomers. Toward this end our laboratory has pursued an electrostatic repulsion approach to the development of potential inhibitors and modulators of hIAPP self-assembly. Peptide conjugates were constructed in which benzene carboxylic acids of varying charge were employed as electrostatic disrupting elements and appended to the N-terminal of the hIAPP22-29 (NFGAILSS) self-recognition sequence. The self-assembly kinetics of conjugates were characterized by turbidity measurements and the structure of aggregates probed by Raman and CD spectroscopy while the morphology was assessed using transmission electron microscopy. Several benzene carboxylic acid peptide conjugates failed to self-assemble and some were found to inhibit the aggregation of full-length amylin while others served to enhance the rate of amyloid formation and/or increase the yield of amyloid produced. Studies reveal that the geometric display of free carboxylates on the benzene ring of the conjugates plays an important role in the activity of conjugates. In addition, a number of free benzene carboxylic acids were found to modulate amylin self-assembly on their own. The results of these investigations confirm the viability of the electrostatic repulsion approach to the modulation of amyloid formation and may aid the design and development of potential therapeutic agents.
Collapse
Affiliation(s)
- Adam A Profit
- Department of Chemistry, York College and The Institute for Macromolecular Assemblies , Jamaica, New York 11451, United States.,Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York , New York, New York 10016, United States
| | - Jayson Vedad
- Department of Chemistry, York College and The Institute for Macromolecular Assemblies , Jamaica, New York 11451, United States.,Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York , New York, New York 10016, United States
| | - Ruel Z B Desamero
- Department of Chemistry, York College and The Institute for Macromolecular Assemblies , Jamaica, New York 11451, United States.,Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York , New York, New York 10016, United States
| |
Collapse
|
21
|
Randino R, Grimaldi M, Persico M, De Santis A, Cini E, Cabri W, Riva A, D’Errico G, Fattorusso C, D’Ursi AM, Rodriquez M. Investigating the Neuroprotective Effects of Turmeric Extract: Structural Interactions of β-Amyloid Peptide with Single Curcuminoids. Sci Rep 2016; 6:38846. [PMID: 28004737 PMCID: PMC5177957 DOI: 10.1038/srep38846] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/15/2016] [Indexed: 02/04/2023] Open
Abstract
A broad biophysical analysis was performed to investigate the molecular basis of the neuroprotective action of Curcuma longa extracts in Alzheimer's disease. By combining circular dichroism and electron paramagnetic resonance experiments with molecular modeling calculations, the minor components of Curcuma longa extracts, such as demethoxycurcumin (2, DMC), bisdemethoxycurcumin (3, BDMC) and cyclocurcumin (4, CYC), were analyzed in a membrane environment mimicking the phospholipid bilayer. Our study provides the first evidence on the relative role of single curcuminoids interacting with Aβ-peptide. When the CYC and curcumin metabolite tetrahydrocurcumin (5, THC) were inserted into an anionic lipid solution, a significant modification of the Aβ CD curves was detected. These data were implemented by EPR experiments, demonstrating that CYC reaches the inner part of the bilayer, while the other curcuminoids are localized close to the membrane interface. Computational studies provided a model for the curcuminoid-Aβ interaction, highlighting the importance of a constrained "semi-folded" conformation to interact with Aβ analogously to the pattern observed in α-helical coiled-coil peptide structures. This combined approach led to a better understanding of the intriguing in vitro and in vivo activity of curcuminoids as anti-Alzheimer agents, paving a new path for the rational design of optimized druggable analogues.
Collapse
Affiliation(s)
- Rosario Randino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084-Fisciano-Italy
| | - Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084-Fisciano-Italy
| | - Marco Persico
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano, 49, 80131-Naples-Italy
| | - Augusta De Santis
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cinthia, 80126-Naples-Italy
| | - Elena Cini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 2, 53100-Siena-Italy
| | - Walter Cabri
- R&D Department, Indena, Viale Ortles, 12, 20139-Milan-Italy
- Innovation & Development Fresenius-Kabi, Piazza Maestri del Lavoro, 7, 20063-Cernusco sul Naviglio Milan-Italy
| | - Antonella Riva
- R&D Department, Indena, Viale Ortles, 12, 20139-Milan-Italy
| | - Gerardino D’Errico
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cinthia, 80126-Naples-Italy
| | - Caterina Fattorusso
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano, 49, 80131-Naples-Italy
| | - Anna Maria D’Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084-Fisciano-Italy
| | - Manuela Rodriquez
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084-Fisciano-Italy
| |
Collapse
|
22
|
|
23
|
Effect of curcumin analogs onα-synuclein aggregation and cytotoxicity. Sci Rep 2016; 6:28511. [PMID: 27338805 PMCID: PMC4919791 DOI: 10.1038/srep28511] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/06/2016] [Indexed: 01/17/2023] Open
Abstract
Alpha-synuclein (α-Syn) aggregation into oligomers and fibrils is associated with dopaminergic neuron loss occurring in Parkinson’s disease (PD) pathogenesis. Compounds that modulate α-Syn aggregation and interact with preformed fibrils/oligomers and convert them to less toxic species could have promising applications in the drug development efforts against PD. Curcumin is one of the Asian food ingredient which showed promising role as therapeutic agent against many neurological disorders including PD. However, the instability and low solubility makes it less attractive for the drug development. In this work, we selected various curcumin analogs and studied their toxicity, stability and efficacy to interact with different α-Syn species and modulation of their toxicity. We found a subset of curcumin analogs with higher stability and showed that curcumin and its various analogs interact with preformed fibrils and oligomers and accelerate α-Syn aggregation to produce morphologically different amyloid fibrils in vitro. Furthermore, these curcumin analogs showed differential binding with the preformed α-Syn aggregates. The present data suggest the potential role of curcumin analogs in modulating α-Syn aggregation.
Collapse
|
24
|
Loureiro JA, Gomes B, Fricker G, Coelho MAN, Rocha S, Pereira MC. Cellular uptake of PLGA nanoparticles targeted with anti-amyloid and anti-transferrin receptor antibodies for Alzheimer's disease treatment. Colloids Surf B Biointerfaces 2016; 145:8-13. [PMID: 27131092 DOI: 10.1016/j.colsurfb.2016.04.041] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 04/12/2016] [Accepted: 04/19/2016] [Indexed: 12/27/2022]
Abstract
During the last few decades, relevant efforts have been reported to design nanocarriers for drug transport through the blood brain barrier (BBB). New drugs, such as peptide iAβ5, capable to inhibit the aggregates associated with Alzheimeŕs disease (AD) are being tested but the most frequent drawback is to reach the brain in the desired concentrations due to the low BBB permeability-surface area. Our approach, as a proof of concept to improve drug transport through the BBB, is based on poly(lactic-co-glycolic acid) (PLGA) nanoparticles with surface functionalized with anti-transferrin receptor monoclonal antibody (OX26) and anti-Aβ (DE2B4) to deliver encapsulated iAβ5 into the brain. Porcine brain capillary endothelial cells (PBCECs) were used as a BBB model to evaluate the system efficacy and toxicity. The uptake of immune nanoparticles with a controlled delivery of the peptide iAβ5 was substantially increased compared to the nanoparticles (NPs) without monoclonal antibody functionalization.
Collapse
Affiliation(s)
- Joana A Loureiro
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Bárbara Gomes
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Gert Fricker
- Institute for Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, 69120 Heidelberg, Germany
| | - Manuel A N Coelho
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Sandra Rocha
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg SE-41296, Sweden
| | - Maria Carmo Pereira
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal.
| |
Collapse
|
25
|
de Almeida NEC, Do TD, Tro M, LaPointe NE, Feinstein SC, Shea JE, Bowers MT. Opposing Effects of Cucurbit[7]uril and 1,2,3,4,6-Penta-O-galloyl-β-d-glucopyranose on Amyloid β25-35 Assembly. ACS Chem Neurosci 2016; 7:218-26. [PMID: 26629788 PMCID: PMC4758880 DOI: 10.1021/acschemneuro.5b00280] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by extracellular deposits of amyloid β protein (Aβ) in the brain. The conversion of soluble monomers to amyloid Aβ fibrils is a complicated process and involves several transient oligomeric species, which are widely believed to be highly toxic and play a crucial role in the etiology of AD. The development of inhibitors to prevent formation of small and midsized oligomers is a promising strategy for AD treatment. In this work, we employ ion mobility spectrometry (IMS), transmission electron microscopy (TEM), and molecular dynamics (MD) simulations to elucidate the structural modulation promoted by two potential inhibitors of Aβ oligomerization, cucurbit[7]uril (CB[7]) and 1,2,3,4,6-penta-O-galloyl-β-d-glucopyranose (PGG), on early oligomer and fibril formation of the Aβ25-35 fragment. One and two CB[7] molecules bind to Aβ25-35 monomers and dimers, respectively, and suppress aggregation by remodeling early oligomer structures and inhibiting the formation of higher-order oligomers. On the other hand, nonselective binding was observed between PGG and Aβ25-35. The interactions between PGG and Aβ25-35, surprisingly, enhanced the formation of Aβ aggregates by promoting extended Aβ25-35 conformations in both homo- and hetero-oligomers. When both ligands were present, the inhibitory effect of CB[7] overrode the stimulatory effect of PGG on Aβ25-35 aggregation, suppressing the formation of large amyloid oligomers and eliminating the structural conversion from isotropic to β-rich topologies induced by PGG. Our results provide mechanistic insights into CB[7] and PGG action on Aβ oligomerization. They also demonstrate the power of the IMS technique to investigate mechanisms of multiple small-molecule agents on the amyloid formation process.
Collapse
Affiliation(s)
- Natália E. C. de Almeida
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Thanh D. Do
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Michael Tro
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Nichole E. LaPointe
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, United States
| | - Stuart C. Feinstein
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, United States
| | - Joan-Emma Shea
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Michael T. Bowers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
- Corresponding author: Michael T. Bowers. Tel: +1-805-893-2673;
| |
Collapse
|
26
|
Liu Y, Wang T, Calabrese AN, Carver JA, Cummins SF, Bowie JH. The membrane-active amphibian peptide caerin 1.8 inhibits fibril formation of amyloid β1-42. Peptides 2015; 73:1-6. [PMID: 26275335 DOI: 10.1016/j.peptides.2015.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/04/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Abstract
The amphibian host-defense peptide caerin 1.8 [(1)GLFKVLGSV(10)AKHLLPHVVP(20)VIAEKL(NH2)] inhibits fibril formation of amyloid β 1-42 [(1)DAEFRHDSG(10)YEVHHQKLVF(20)FAEDVGSNKG(30)AIIGLMVGGV(40)VIA] [Aβ42] (the major precursor of the extracellular fibrillar deposits of Alzheimer's disease). Some truncated forms of caerin 1.8 also inhibit fibril formation of Aβ42. For example, caerin 1.8 (1-13) [(1)GLFKVLGSV(10)AKHL(NH2) and caerin 1.8 (22-25) [KVLGSV(10)AKHLLPHVVP(20)VIAEKL(NH2)] show 85% and 75% respectively of the inhibition activity of the parent caerin 1.8. The synthetic peptide KLVFFKKKKKK is a known inhibitor of Aβ42 fibril formation, and was used as a standard in this study. Caerin 1.8 is the more effective fibril inhibitor. IC50 values (± 15%) are caerin 1.8 (75 μM) and KLVFFKKKKKK (370 μM). MALDI mass spectrometry shows the presence of a small peak corresponding to a protonated 1:1 adduct [caerin 1.8/Aβ42]H(+). Molecular dynamics simulation suggests that both hydrogen bonding and hydrophobic interactions between Aβ42 and caerin 1.8 facilitate the formation of a 1:1 complex in water. Fibril formation from Aβ42 has been proposed to be based around the (16)KLVF(20)F region of Aβ42; this region in the 1:1 complex is partially blocked from attachment of a further molecule of Aβ42.
Collapse
Affiliation(s)
- Yanqin Liu
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia
| | - Tianfang Wang
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, Queensland 4558, Australia
| | - Antonio N Calabrese
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia
| | - John A Carver
- Research School of Chemistry, The Australian National University, Canberra, Australian Capital Chemistry, 2601, Australia
| | - Scott F Cummins
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, Queensland 4558, Australia
| | - John H Bowie
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia.
| |
Collapse
|
27
|
Di Carlo MG, Minicozzi V, Foderà V, Militello V, Vetri V, Morante S, Leone M. Thioflavin T templates amyloid β(1–40) conformation and aggregation pathway. Biophys Chem 2015; 206:1-11. [DOI: 10.1016/j.bpc.2015.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/03/2015] [Accepted: 06/09/2015] [Indexed: 12/31/2022]
|
28
|
Biflavonoids as Potential Small Molecule Therapeutics for Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 863:55-77. [PMID: 26092626 DOI: 10.1007/978-3-319-18365-7_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Flavonoids are naturally occurring phytochemicals found in a variety of fruits and vegetables and offer color, flavor, aroma, nutritional and health benefits. Flavonoids have been found to play a neuroprotective role by inhibiting and/or modifying the self-assembly of the amyloid-β (Aβ) peptide into oligomers and fibrils, which are linked to the pathogenesis of Alzheimer's disease. The neuroprotective efficacy of flavonoids has been found to strongly depend on their structure and functional groups. Flavonoids may exist in monomeric, as well as di-, tri-, tetra- or polymeric form through C-C or C-O-C linkages. It has been shown that flavonoids containing two or more units, e.g., biflavonoids, exert greater biological activity than their respective monoflavonoids. For instance, biflavonoids have the ability to distinctly alter Aβ aggregation and more effectively reduce the toxicity of Aβ oligomers compared to the monoflavonoid moieties. Although the molecular mechanisms remain to be elucidated, flavonoids have been shown to alter the Aβ aggregation pathway to yield non-toxic, unstructured Aβ aggregates, as well as directly exerting a neuroprotective effect to cells. In this chapter, we review biflavonoid-mediated Aβ aggregation and toxicity, and highlight the beneficial roles biflavonoids can potentially play in the prevention and treatment of Alzheimer's disease.
Collapse
|
29
|
|
30
|
Hernández-Rodríguez M, Correa-Basurto J, Nicolás-Vázquez MI, Miranda-Ruvalcaba R, Benítez-Cardoza CG, Reséndiz-Albor AA, Méndez-Méndez JV, Rosales-Hernández MC. Virtual and In Vitro Screens Reveal a Potential Pharmacophore that Avoids the Fibrillization of Aβ1-42. PLoS One 2015; 10:e0130263. [PMID: 26172152 PMCID: PMC4501547 DOI: 10.1371/journal.pone.0130263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/18/2015] [Indexed: 11/25/2022] Open
Abstract
Among the multiple factors that induce Alzheimer’s disease, aggregation of the amyloid β peptide (Aβ) is considered the most important due to the ability of the 42-amino acid Aβ peptides (Aβ1–42) to form oligomers and fibrils, which constitute Aβ pathological aggregates. For this reason, the development of inhibitors of Aβ1–42 pathological aggregation represents a field of research interest. Several Aβ1–42 fibrillization inhibitors possess tertiary amine and aromatic moieties. In the present study, we selected 26 compounds containing tertiary amine and aromatic moieties with or without substituents and performed theoretical studies that allowed us to select four compounds according to their free energy values for Aβ1–42 in α-helix (Aβ-α), random coil (Aβ-RC) and β-sheet (Aβ-β) conformations. Docking studies revealed that compound 5 had a higher affinity for Aβ-α and Aβ-RC than the other compounds. In vitro, this compound was able to abolish Thioflavin T fluorescence and favored an RC conformation of Aβ1–42 in circular dichroism studies, resulting in the formation of amorphous aggregates as shown by atomic force microscopy. The results obtained from quantum studies allowed us to identify a possible pharmacophore that can be used to design Aβ1–42 aggregation inhibitors. In conclusion, compounds with higher affinity for Aβ-α and Aβ-RC prevented the formation of oligomeric species.
Collapse
Affiliation(s)
- Maricarmen Hernández-Rodríguez
- Laboratorio de Modelado Molecular y Diseño de Fármacos, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Delegación Miguel Hidalgo, México D.F., México
- Laboratorio de Biofísica y Biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Delegación Miguel Hidalgo, México D.F., México
| | - José Correa-Basurto
- Laboratorio de Modelado Molecular y Diseño de Fármacos, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Delegación Miguel Hidalgo, México D.F., México
- * E-mail: (MCRH): (JCB)
| | - María Inés Nicolás-Vázquez
- Quimica inorgánica-orgánica del Departamento de Ciencias Químicas, de la Facultad de Estudios Superiores Cuautitlán Campo 1, Universidad Nacional Autónoma de México, Avenida 1o de Mayo S/N, Santa María las Torres, Cuautitlán Izcalli, Estado de México, México
| | - René Miranda-Ruvalcaba
- Quimica inorgánica-orgánica del Departamento de Ciencias Químicas, de la Facultad de Estudios Superiores Cuautitlán Campo 1, Universidad Nacional Autónoma de México, Avenida 1o de Mayo S/N, Santa María las Torres, Cuautitlán Izcalli, Estado de México, México
| | - Claudia Guadalupe Benítez-Cardoza
- Laboratorio de Investigación Bioquímica, Sección de Estudios de Posgrado e Investigación, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu H 239, Gustavo A. Madero, La Escalera, México D.F., México
| | - Aldo Arturo Reséndiz-Albor
- Laboratorio de Investigación en Inmunología., Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Delegación Miguel Hidalgo, México D.F., México
| | - Juan Vicente Méndez-Méndez
- Centro de Nanociencias y Micro y Nanotecnología, Instituto Politécnico Nacional, Luis Enrique Erro S/N, U. Prof Adolfo López Mateos, Gustavo A. Madero, México D.F., México
| | - Martha C. Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Delegación Miguel Hidalgo, México D.F., México
- * E-mail: (MCRH): (JCB)
| |
Collapse
|
31
|
Schor M, Mey ASJS, Noé F, MacPhee CE. Shedding Light on the Dock-Lock Mechanism in Amyloid Fibril Growth Using Markov State Models. J Phys Chem Lett 2015; 6:1076-1081. [PMID: 26262873 DOI: 10.1021/acs.jpclett.5b00330] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
We investigate how the molecular mechanism of monomer addition to a growing amyloid fibril of the transthyretin TTR105-115 peptide is affected by pH. Using Markov state models to extract equilibrium and dynamical information from extensive all atom simulations allowed us to characterize both productive pathways in monomer addition as well as several off-pathway trapped states. We found that multiple pathways result in successful addition. All productive pathways are driven by the central hydrophobic residues in the peptide. Furthermore, we show that the slowest transitions in the system involve trapped configurations, that is, long-lived metastable states. These traps dominate the rate of fibril growth. Changing the pH essentially reweights the system, leading to clear differences in the relative importance of both productive paths and traps, yet retains the core mechanism.
Collapse
Affiliation(s)
- Marieke Schor
- †School of Physics and Astronomy, University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, United Kingdom
| | - Antonia S J S Mey
- ‡Department for Mathematics and Computer Science, Freie Universität Berlin, Arnimallee 6, Berlin 14195, Germany
| | - Frank Noé
- ‡Department for Mathematics and Computer Science, Freie Universität Berlin, Arnimallee 6, Berlin 14195, Germany
| | - Cait E MacPhee
- †School of Physics and Astronomy, University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, United Kingdom
| |
Collapse
|
32
|
Bhatia NK, Srivastava A, Katyal N, Jain N, Khan MAI, Kundu B, Deep S. Curcumin binds to the pre-fibrillar aggregates of Cu/Zn superoxide dismutase (SOD1) and alters its amyloidogenic pathway resulting in reduced cytotoxicity. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:426-36. [PMID: 25666897 DOI: 10.1016/j.bbapap.2015.01.014] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 12/25/2014] [Accepted: 01/31/2015] [Indexed: 12/13/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease that affects motor neurons. Unfortunately, effective therapeutics against this disease is still not available. Almost 20% of familial ALS (fALS) is suggested to be associated with pathological deposition of superoxide dismutase (SOD1). Evidences suggest that SOD1-containing pathological inclusions in ALS exhibit amyloid like properties. An effective strategy to combat ALS may be to inhibit amyloid formation of SOD1 using small molecules. In the present study, we observed the fibrillation of one of the premature forms of SOD1 (SOD1 with reduced disulfide) in the presence of curcumin. Using ThT binding assay, AFM, TEM images and FTIR, we demonstrate that curcumin inhibits the DTT-induced fibrillation of SOD1 and favors the formation of smaller and disordered aggregates of SOD1. The enhancement in curcumin fluorescence on the addition of oligomers and pre-fibrillar aggregates of SOD1 suggests binding of these species to curcumin. Docking studies indicate that putative binding site of curcumin may be the amyloidogenic regions of SOD1. Further, there is a significant increase in SOD1 mediated toxicity in the regime of pre-fibrillar and fibrillar aggregates which is not evident in curcumin containing samples. All these data suggest that curcumin reduces toxicity by binding to the amyloidogenic regions of the species on the aggregation pathway and blocking the formation of the toxic species. Nanoparticles of curcumin with higher aqueous solubility show similar aggregation control as that of curcumin bulk. This suggests a potential role for curcumin in the treatment of ALS.
Collapse
Affiliation(s)
- Nidhi K Bhatia
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, India
| | - Ankit Srivastava
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, India
| | - Nidhi Katyal
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, India
| | - Nidhi Jain
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, India
| | - M Ashhar I Khan
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, India
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, India.
| |
Collapse
|
33
|
Patil SM, Alexandrescu AT. Charge-Based Inhibitors of Amylin Fibrillization and Toxicity. J Diabetes Res 2015; 2015:946037. [PMID: 26576438 PMCID: PMC4630399 DOI: 10.1155/2015/946037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/12/2014] [Indexed: 02/01/2023] Open
Abstract
To test the hypothesis that electrostatic repulsion is an important force opposing amyloid fibril assembly, we designed peptides that substitute strings of positively or negatively charged residues into the sequence of the amyloidogenic hormone amylin, which contributes to type 2 diabetes pathology. Arg-1 and Arg-2 substitute four positively charged arginines for segments that in structural models of amylin fibrils form the end of strand β1 and the beginning of strand β2, respectively. Mem-T substitutes negatively charged aspartates for the peptide segment with the largest avidity for membranes. All three charge-loaded peptides fibrillize poorly on their own and inhibit fibril elongation of WT-amylin at physiological ionic strength. The inhibition of WT-amylin fibril elongation rates is salt-dependent indicating that the analogs act through electrostatic interactions. Arg-1 protects against WT-amylin cytotoxicity towards a MIN6 mouse model of pancreatic β-cells, and Arg-2 protects at higher concentrations, whereas Mem-T has no effect. The most effective variant, Arg-1, inhibits WT-amylin fibril elongation rates with an IC50 of ~1 µM and cytotoxicity with an IC50 of ~50 µM, comparable to other types of fibrillization inhibitors reported in the literature. Taken together, these results suggest that electrostatic interactions can be exploited to develop new types of inhibitors of amyloid fibrillization and toxicity.
Collapse
Affiliation(s)
- Sharadrao M. Patil
- Department of Molecular and Cell Biology, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269-3125, USA
| | - Andrei T. Alexandrescu
- Department of Molecular and Cell Biology, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269-3125, USA
- *Andrei T. Alexandrescu:
| |
Collapse
|
34
|
Babu E, Muthu Mareeswaran P, Sathish V, Singaravadivel S, Rajagopal S. Sensing and inhibition of amyloid-β based on the simple luminescent aptamer-ruthenium complex system. Talanta 2014; 134:348-353. [PMID: 25618678 DOI: 10.1016/j.talanta.2014.11.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/08/2014] [Accepted: 11/10/2014] [Indexed: 12/21/2022]
Abstract
Aggregation of amyloid-β (Aβ) peptide has been known to be pathologically associated with Alzheimer and dementia diseases. Amyloid-β fibrils serve as an important target for the drugs development and diagnosis of neurodegenerative diseases. Herein, we report a new [Ru(dmbpy)(dcbpy)dppz)] complex (dmbpy; 4,4'-dimethyl-2,2'-bipyridine, dcbpy; 4,4'-dicorboxy-2,2'-bipyridine, dppz; dipyridophenazine) intercalated aptamer based recognition of amyloid-β. Interestingly, aforementioned Ru(II) complex shows weak luminescence intensity in the aqueous medium but it shows strong luminescence intensity in the presence of RNA aptamer. Upon addition of amyloid-β monomers, the luminescence intensity of Ru(II) complex is reduced due to the strong interaction of aptamer with amyloid-β monomer/small oligomers. Furthermore, present study implies that our system has ability to inhibit the formation of amyloid-β fibrils, which is confirmed from the AFM morphological structures in the absence and presence of aptamer. This work may contribute the rapid diagnosis and inhibition of amyloid-β aggregation in the clinical applications.
Collapse
Affiliation(s)
- Eththilu Babu
- Department of Physical Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, Tamil Nadu, India; Department of Chemistry, VV College of Engineering, Tisaiyanvilai, Tamil Nadu, India
| | - Paulpandian Muthu Mareeswaran
- Department of Physical Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, Tamil Nadu, India; Graduate School of EEWS, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Veerasamy Sathish
- Department of Physical Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, Tamil Nadu, India; Department of Physical Science, Bannari Amman Institute of Technology, Sathiyamangalam, Tamil Nadu, India
| | - Subramanian Singaravadivel
- Department of Physical Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Seenivasan Rajagopal
- Department of Physical Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, Tamil Nadu, India.
| |
Collapse
|
35
|
Li J, Han Q, Wang X, Yu N, Yang L, Yang R, Wang C. Reduced aggregation and cytotoxicity of amyloid peptides by graphene oxide/gold nanocomposites prepared by pulsed laser ablation in water. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:4386-4394. [PMID: 25059878 DOI: 10.1002/smll.201401121] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/18/2014] [Indexed: 06/03/2023]
Abstract
A novel and convenient method to synthesize the nanocomposites combining graphene oxides (GO) with gold nanoparticles (AuNPs) is reported and their applications to modulate amyloid peptide aggregation are demonstrated. The nanocomposites produced by pulsed laser ablation (PLA) in water show good biocompatibility and solubility. The reduced aggregation of amyloid peptides by the nanocomposites is confirmed by Thioflavin T fluorescence and atomic force microscopy. The cell viability experiments reveals that the presence of the nanocomposites can significantly reduce the cytotoxicity of the amyloid peptides. Furthermore, the depolymerization of peptide fibrils and inhibition of their cellular cytotoxicity by GO/AuNPs is also observed. These observations suggest that the nanocomposites combining GO and AuNPs have a great potential for designing new therapeutic agents and are promising for future treatment of amyloid-related diseases.
Collapse
Affiliation(s)
- Jingying Li
- CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, (China)
| | | | | | | | | | | | | |
Collapse
|
36
|
Antimisiaris S, Mourtas S, Markoutsa E, Skouras A, Papadia K. Nanoparticles for Diagnosis and/or Treatment of Alzheimer's Disease. Adv Healthc Mater 2014. [DOI: 10.1002/9781118774205.ch4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
37
|
Chinisaz M, Ghasemi A, Larijani B, Ebrahim-Habibi A. Amyloid formation and inhibition of an all-beta protein: A study on fungal polygalacturonase. J Mol Struct 2014. [DOI: 10.1016/j.molstruc.2013.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Honcharenko D, Bose PP, Maity J, Kurudenkandy FR, Juneja A, Flöistrup E, Biverstål H, Johansson J, Nilsson L, Fisahn A, Strömberg R. Synthesis and evaluation of antineurotoxicity properties of an amyloid-β peptide targeting ligand containing a triamino acid. Org Biomol Chem 2014; 12:6684-93. [DOI: 10.1039/c4ob00959b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new triamino acid enables synthesis of an amyloid-β peptide (Aβ) targeting ligand with additional Aβ–ligand interactions that gives protection towards Aβ-induced reduction of gamma oscillations in hippocampal slice preparation.
Collapse
Affiliation(s)
- Dmytro Honcharenko
- Department of Biosciences and Nutrition
- Karolinska Institutet
- S-14183 Huddinge, Sweden
| | - Partha Pratim Bose
- Department of Biosciences and Nutrition
- Karolinska Institutet
- S-14183 Huddinge, Sweden
| | - Jyotirmoy Maity
- Department of Biosciences and Nutrition
- Karolinska Institutet
- S-14183 Huddinge, Sweden
| | - Firoz Roshan Kurudenkandy
- Neuronal Oscillations Laboratory
- Division for Neurogeriatrics
- KI-Alzheimer Disease Research Center
- Department of Neurobiology
- Care Sciences and Society
| | - Alok Juneja
- Department of Biosciences and Nutrition
- Karolinska Institutet
- S-14183 Huddinge, Sweden
| | - Erik Flöistrup
- Department of Biosciences and Nutrition
- Karolinska Institutet
- S-14183 Huddinge, Sweden
| | - Henrik Biverstål
- KI-Alzheimer Disease Research Center
- Department of Neurobiology
- Care Sciences and Society
- Karolinska Institute
- 14186 Huddinge, Sweden
| | - Jan Johansson
- KI-Alzheimer Disease Research Center
- Department of Neurobiology
- Care Sciences and Society
- Karolinska Institute
- 14186 Huddinge, Sweden
| | - Lennart Nilsson
- Department of Biosciences and Nutrition
- Karolinska Institutet
- S-14183 Huddinge, Sweden
| | - André Fisahn
- Neuronal Oscillations Laboratory
- Division for Neurogeriatrics
- KI-Alzheimer Disease Research Center
- Department of Neurobiology
- Care Sciences and Society
| | - Roger Strömberg
- Department of Biosciences and Nutrition
- Karolinska Institutet
- S-14183 Huddinge, Sweden
| |
Collapse
|
39
|
Bleiholder C, Do TD, Wu C, Economou NJ, Bernstein SS, Buratto SK, Shea JE, Bowers MT. Ion mobility spectrometry reveals the mechanism of amyloid formation of Aβ(25-35) and its modulation by inhibitors at the molecular level: epigallocatechin gallate and scyllo-inositol. J Am Chem Soc 2013; 135:16926-37. [PMID: 24131107 DOI: 10.1021/ja406197f] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Amyloid cascades leading to peptide β-sheet fibrils and plaques are central to many important diseases. Recently, intermediate assemblies of these cascades were identified as the toxic agents that interact with the cellular machinery. The relationship between the transformation from natively unstructured assembly to the β-sheet oligomers to disease is important in understanding disease onset and the development of therapeutic agents. Research on this early oligomeric region has largely been unsuccessful since traditional techniques measure only ensemble average oligomer properties. Here, ion mobility methods are utilized to deduce the modulation of peptide self-assembly pathways in the amyloid-β protein fragment Aβ(25-35) by two amyloid inhibitors (epigallocatechin gallate and scyllo-inositol) that are currently in clinical trials for Alzheimer's Disease. We provide evidence that suppression of β-extended oligomers from the onset of the conversion into β-oligomer conformations is essential for effective attenuation of β-structured amyloid oligomeric species often associated with oligomer toxicity. Furthermore, we demonstrate the ease with which ion mobility spectrometry-mass spectrometry can guide the development of therapeutic agents and drug evaluation by providing molecular level insight into the amyloid formation process and its modulation by small molecule assembly modulators.
Collapse
Affiliation(s)
- Christian Bleiholder
- Department of Chemistry and Biochemistry, University of California , Santa Barbara, California 93106-9510, United States
| | | | | | | | | | | | | | | |
Collapse
|
40
|
He Y, Yao PF, Chen SB, Huang ZH, Huang SL, Tan JH, Li D, Gu LQ, Huang ZS. Synthesis and evaluation of 7,8-dehydrorutaecarpine derivatives as potential multifunctional agents for the treatment of Alzheimer's disease. Eur J Med Chem 2013; 63:299-312. [DOI: 10.1016/j.ejmech.2013.02.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/31/2013] [Accepted: 02/13/2013] [Indexed: 10/27/2022]
|
41
|
Okamoto A, Nomura K, Yano A, Higai S, Kondo T, Kamba S, Kurita N. Proposal for an inhibitor of Alzheimer's disease blocking aggregation of amyloid-β peptides:ab initiomolecular simulations. ACTA ACUST UNITED AC 2013. [DOI: 10.1088/1742-6596/433/1/012033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
42
|
Singh PK, Kotia V, Ghosh D, Mohite GM, Kumar A, Maji SK. Curcumin modulates α-synuclein aggregation and toxicity. ACS Chem Neurosci 2013; 4:393-407. [PMID: 23509976 DOI: 10.1021/cn3001203] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In human beings, Parkinson's disease (PD) is associated with the oligomerization and amyloid formation of α-synuclein (α-Syn). The polyphenolic Asian food ingredient curcumin has proven to be effective against a wide range of human diseases including cancers and neurological disorders. While curcumin has been shown to significantly reduce cell toxicity of α-Syn aggregates, its mechanism of action remains unexplored. Here, using a series of biophysical techniques, we demonstrate that curcumin reduces toxicity by binding to preformed oligomers and fibrils and altering their hydrophobic surface exposure. Further, our fluorescence and two-dimensional nuclear magnetic resonance (2D-NMR) data indicate that curcumin does not bind to monomeric α-Syn but binds specifically to oligomeric intermediates. The degree of curcumin binding correlates with the extent of α-Syn oligomerization, suggesting that the ordered structure of protein is required for effective curcumin binding. The acceleration of aggregation by curcumin may decrease the population of toxic oligomeric intermediates of α-Syn. Collectively; our results suggest that curcumin and related polyphenolic compounds can be pursued as candidate drug targets for treatment of PD and other neurological diseases.
Collapse
Affiliation(s)
- Pradeep K. Singh
- Department of Biosciences and Bioengineering and ‡Wadhwani Research
Centre for Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra,
India 400076
| | - Vasudha Kotia
- Department of Biosciences and Bioengineering and ‡Wadhwani Research
Centre for Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra,
India 400076
| | - Dhiman Ghosh
- Department of Biosciences and Bioengineering and ‡Wadhwani Research
Centre for Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra,
India 400076
| | - Ganesh M. Mohite
- Department of Biosciences and Bioengineering and ‡Wadhwani Research
Centre for Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra,
India 400076
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering and ‡Wadhwani Research
Centre for Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra,
India 400076
| | - Samir K. Maji
- Department of Biosciences and Bioengineering and ‡Wadhwani Research
Centre for Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra,
India 400076
| |
Collapse
|
43
|
Bleiholder C, Dupuis NF, Bowers MT. Dimerization of Chirally Mutated Enkephalin Neurotransmitters: Implications for Peptide and Protein Aggregation Mechanisms. J Phys Chem B 2013; 117:1770-9. [DOI: 10.1021/jp306386p] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Christian Bleiholder
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California
93106, United States
| | - Nicholas F. Dupuis
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California
93106, United States
| | - Michael T. Bowers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California
93106, United States
| |
Collapse
|
44
|
Antosova A, Gazova Z, Fedunova D, Valusova E, Bystrenova E, Valle F, Daxnerova Z, Biscarini F, Antalik M. Anti-amyloidogenic activity of glutathione-covered gold nanoparticles. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2012. [DOI: 10.1016/j.msec.2012.07.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
45
|
A multifunctional peptide rescues memory deficits in Alzheimer's disease transgenic mice by inhibiting Aβ42-induced cytotoxicity and increasing microglial phagocytosis. Neurobiol Dis 2012; 46:701-9. [DOI: 10.1016/j.nbd.2012.03.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 02/20/2012] [Accepted: 03/01/2012] [Indexed: 12/25/2022] Open
|
46
|
Effects of ligands on unfolding of the amyloid β-peptide central helix: mechanistic insights from molecular dynamics simulations. PLoS One 2012; 7:e30510. [PMID: 22291970 PMCID: PMC3264620 DOI: 10.1371/journal.pone.0030510] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 12/22/2011] [Indexed: 11/28/2022] Open
Abstract
Polymerization of the amyloid β-peptide (Aβ), a process which requires that the helical structure of Aβ unfolds beforehand, is suspected to cause neurodegeneration in Alzheimer's disease. According to recent experimental studies, stabilization of the Aβ central helix counteracts Aβ polymerization into toxic assemblies. The effects of two ligands (Dec-DETA and Pep1b), which were designed to bind to and stabilize the Aβ central helix, on unfolding of the Aβ central helix were investigated by molecular dynamics simulations. It was quantitatively demonstrated that the stability of the Aβ central helix is increased by both ligands, and more effectively by Pep1b than by Dec-DETA. In addition, it was shown that Dec-DETA forms parallel conformations with β-strand-like Aβ, whereas Pep1b does not and instead tends to bend unwound Aβ. The molecular dynamics results correlate well with previous experiments for these ligands, which suggest that the simulation method should be useful in predicting the effectiveness of novel ligands in stabilizing the Aβ central helix. Detailed Aβ structural changes upon loss of helicity in the presence of the ligands are also revealed, which gives further insight into which ligand may lead to which path subsequent to unwinding of the Aβ central helix.
Collapse
|
47
|
Brandenburg E, von Berlepsch H, Gerling UIM, Böttcher C, Koksch B. Inhibition of amyloid aggregation by formation of helical assemblies. Chemistry 2012; 17:10651-61. [PMID: 22003512 DOI: 10.1002/chem.201100670] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The formation of amyloid aggregates is responsible for a wide range of diseases, including Alzheimer's and Parkinson's disease. Although the amyloid-forming proteins have different structures and sequences, all undergo a conformational change to form amyloid aggregates that have a characteristic cross-β-structure. The mechanistic details of this process are poorly understood, but different strategies for the development of inhibitors of amyloid formation have been proposed. In most cases, chemically diverse compounds bind to an elongated form of the protein in a β-strand conformation and thereby exert their therapeutic effect. However, this approach could favor the formation of prefibrillar oligomeric species, which are thought to be toxic. Herein, we report an alternative approach in which a helical coiled-coil-based inhibitor peptide has been designed to engage a coiled-coil-based amyloid-forming model peptide in a stable coiled-coil arrangement, thereby preventing rearrangement into a β-sheet conformation and the subsequent formation of amyloid-like fibrils. Moreover, we show that the helix-forming peptide is able to disassemble mature amyloid-like fibrils.
Collapse
|
48
|
Zou Z, Fan Y, Zhang C. Preventing protein aggregation by its hyper-acidic fusion cognates in Escherichia coli. Protein Expr Purif 2011; 80:138-44. [DOI: 10.1016/j.pep.2011.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 06/06/2011] [Accepted: 06/07/2011] [Indexed: 12/31/2022]
|
49
|
Liu FF, Dong XY, He L, Middelberg APJ, Sun Y. Molecular insight into conformational transition of amyloid β-peptide 42 inhibited by (-)-epigallocatechin-3-gallate probed by molecular simulations. J Phys Chem B 2011; 115:11879-87. [PMID: 21899367 DOI: 10.1021/jp202640b] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Considerable experimental evidence indicates that (-)-epigallocatechin-3-gallate (EGCG) inhibits the fibrillogenesis of Aβ(42) and alleviates its associated cytotoxicity. However, the molecular mechanism of the inhibition effect of EGCG on the conformational transition of Aβ(42) remains unclear due to the limitations of current experimental techniques. In this work, molecular dynamics simulations and molecular mechanics-Poisson-Boltzmann surface area (MM-PBSA) analysis were coupled to better understand the issue. It was found that the direct interactions between EGCG and the peptide are the origin of its inhibition effects. Specifically, EGCG molecules expel water from the surface of the Aβ(42), cluster with each other, and interact directly with the peptide. The results of free energy decomposition calculated by MM-PBSA indicate that the nonpolar term contributes more than 71% to the binding free energy of the EGCG-Aβ(42) complex, while polar interactions (i.e., hydrogen bonding) play a minor role. It was identified that there are 12 important residues of Aβ(42) that strongly interact with EGCG (Phe4, Arg5, Phe19, Phe20, Glu22, Lys28, Gly29, Leu34-Gly37, and Ile41), while nonpolar interactions are mainly provided by the side chains of some hydrophobic residues (Phe, Met and Ile) and the main chains of some nonhydrophobic residues (Lys28 and Gly29). On the contrary, polar interactions are mainly formed by the main chain of Aβ(42), of which the main chains of Gly29 and Gly37 contribute greatly. The work has thus elucidated the molecular mechanism of the inhibition effect of EGCG on the conformational transition of Aβ(42), and the findings are considered critical for exploring more effective agents for the inhibition of Aβ(42) fibrillogenesis.
Collapse
Affiliation(s)
- Fu-Feng Liu
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | | | | | | | | |
Collapse
|
50
|
Design and development of peptides and peptide mimetics as antagonists for therapeutic intervention. Future Med Chem 2011; 2:1813-22. [PMID: 21428804 DOI: 10.4155/fmc.10.259] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The concept of peptides as therapeutic agents has been historically disregarded by the pharmaceutical industry on account of their susceptibility to degradation, their size and consequent limitations in methods of delivery. Recently, however, there has been a surge of interest in peptides and their mimetics as potential antagonists for therapeutic intervention. This is in part due to the increased half-life and oral availability that has been achieved for a number of peptide-based systems, the introduction and acceptance of alternative delivery methods, and the prevalence of proteomics to identify countless protein-protein interaction targets. The use of peptides and molecules that mimic their function therefore has great potential to effectively target a range of proteins that are pathogenically implicated in numerous diseases.
Collapse
|