1
|
Deng J, Wan W, Sun R, Xia Q, Yan J, Sun J, Jia X, Jin H, Wang X, Guo K, Li M, Liu Y. Acid-Resistant and Viscosity-Sensitive Proteome Aggregation Sensor To Visualize Cellular Aggrephagy in Live Cells and Clinical Samples. ACS Sens 2025; 10:2812-2822. [PMID: 40189840 DOI: 10.1021/acssensors.4c03560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Aggrephagy in cells is defined as the degradation of intracellular aggregated proteins via the macroautophagy process. This process sequesters protein aggregates into autolysosomes, which bear characteristic viscous and acidic microenvironments. Limited protein aggregation sensors are environmentally compatible with the cellular aggrephagy process. Here, we report an acid-resistant and viscosity-sensitive proteome aggregation sensor to detect cellular aggrephagy in stressed cells and clinical samples. This sensor fluoresces upon selectively and ubiquitously binding to different aggregated proteins. Importantly, unlike other reported protein aggregation sensors, our probe offers unique acid-resistant fluorescence inside aggregated proteins, enabling its application in the acidic autolysosome microenvironment. In live cells under various stressed conditions, the optimal probe (A6) successfully detects aggregated proteome in autolysosomes, as validated by colocalization with a lysosomal tracker. Additionally, we demonstrate that the sensor can detect proteome aggregation in heat-stressed clinical tissue samples biopsied from cancer patients undergoing thermal perfusion treatment. Together, the reported acid-resistant and viscosity-sensitive protein aggregation sensor facilitates the detection of cellular aggrephagy by chemically matching its microenvironmental characteristics.
Collapse
Affiliation(s)
- Jintai Deng
- The Second Hospital of Dalian Medical University, Dalian 116023, China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Wang Wan
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui Sun
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiuxuan Xia
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Yan
- The Second Hospital of Dalian Medical University, Dalian 116023, China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Jialu Sun
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Xiaomeng Jia
- The Second Hospital of Dalian Medical University, Dalian 116023, China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Hao Jin
- The Second Hospital of Dalian Medical University, Dalian 116023, China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Xueqing Wang
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Kun Guo
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Man Li
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Yu Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
2
|
La Manna S, Panzetta V, Graziano SF, Cipollone I, Tolbatov I, Marrone A, Monti M, Netti PA, Merlino A, Kowalski K, Marasco D. Heterobimetallic ferrocenyl-chromone compounds as selective inhibitors of amyloid aggregation. J Inorg Biochem 2025; 270:112932. [PMID: 40288002 DOI: 10.1016/j.jinorgbio.2025.112932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Amyloid aggregation is a key process in neurodegeneration, producing toxic species that contribute to disease progression. This underscores the urgent need to identify novel agents capable of reducing toxicity by modulating this aggregation process. Two heterobimetallic complexes incorporating a ferrocenyl-chromone (Fc-Chr) core were investigated: one featuring an additional gold(I) triphenylphosphine moiety, Fc-Chr-AuP(Ph)₃, and the other containing a dicobalt hexacarbonyl-alkyne unit, Fc-Chr-Co₂(CO)₆. Their effects were evaluated toward on the aggregation of two amyloid models: the peptide spanning residues 264-277 of nucleophosmin 1 (NPM1264-277) and the C-terminal fragment of the amyloid-β peptide (Aβ21-40) each with unique primary sequences, self-aggregation mechanisms and kinetics. Thioflavin T- assays allowed to assess the impact of the metal complexes on the aggregation of two amyloids. Results indicate that the two complexes inhibit the early stages of the aggregation of peptides in a dose-dependent manner and a greater effect on NPM1264-277 when compared to Aβ21-40 was observed. Native electrospray ionization mass spectrometry revealed the formation of peculiar metal/peptide adducts in dependence on different metal-units in the complexes. Scanning electron microscopy (SEM) and density function theory (DFT) were also employed to further characterize the interaction between the metal compounds and the investigated peptides, while preliminary cell viability assays in SH-SY5Y cells supported inhibitory effects on the aggregation and showed reduction of amyloid cytotoxicity. Fc-Chr-Co₂(CO)₆ demonstrated the highest efficacy in modulating peptide aggregation, exerting a more significant impact on NMP1264-277 relative to Aβ₂₁₋₄₀. These results support the use of ferrocenyl-chromone containing metal complexes as modulators of amyloid peptide aggregation.
Collapse
Affiliation(s)
- Sara La Manna
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Valeria Panzetta
- Department of Ingegneria Chimica del Materiali e della Produzione Industriale (DICMAPI), University of Naples Federico II, 80125 Naples, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | | | - Irene Cipollone
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Iogann Tolbatov
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, 07100 Sassari, Italy
| | - Alessandro Marrone
- Department of Pharmacy, University of Chieti-Pescara "G. d' Annunzio", 66100 Chieti, Italy
| | - Maria Monti
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Paolo Antonio Netti
- Department of Ingegneria Chimica del Materiali e della Produzione Industriale (DICMAPI), University of Naples Federico II, 80125 Naples, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Konrad Kowalski
- University of Łódź, Faculty of Chemistry, Department of Organic Chemistry, Tamka 12, 91-403 Łódź, Poland
| | - Daniela Marasco
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
| |
Collapse
|
3
|
Günnaz S, Yildiz E, Tunçel Oral A, Yurt F, Erdem A, Irişli S. Schiff Base-platinum and ruthenium complexes and anti-Alzheimer properties. J Inorg Biochem 2025; 264:112790. [PMID: 39626364 DOI: 10.1016/j.jinorgbio.2024.112790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/15/2024] [Accepted: 11/24/2024] [Indexed: 01/12/2025]
Abstract
This study investigates the effects of Pt and Ru complexes containing a Schiff base with a diimine structure on Alzheimer's disease. The Schiff base (N1E,N2E)-N1,N2-bis(isoquinolin-4-ylmethylene)benzene-1,2-diamine (I) and the novel Pt(II) and Ru(II) complexes (Ia and Ib) were synthesized and characterized using FTIR, NMR (1H, 13C), mass spectrometry, and elemental analyses. Their ability to inhibit amyloid beta (Aβ1-42) aggregation was determined in vitro using the SH-SY5Y cell line. Fluorescence spectroscopy investigated the early aggregation kinetics and dose-dependent characteristics of Aβ1-42 with the complexes. Transmission electron microscopy confirmed the results. Matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) and 1H NMR spectroscopy examined the interaction with Aβ1-16. Electrochemical analysis using square wave voltammetry monitored the interaction with Aβ1-42. The synthesized complexes were active in inhibiting amyloid aggregation at a low molar ratio.
Collapse
Affiliation(s)
- Salih Günnaz
- Department of Chemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| | - Esma Yildiz
- Department of Analytical Chemistry, Faculty of Pharmacy, Ege University, Bornova, Izmir 35100, Turkey
| | - Ayça Tunçel Oral
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Fatma Yurt
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Arzum Erdem
- Department of Analytical Chemistry, Faculty of Pharmacy, Ege University, Bornova, Izmir 35100, Turkey
| | - Sevil Irişli
- Department of Chemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey.
| |
Collapse
|
4
|
Ghosh P, Kundu A, Ganguly D. From experimental studies to computational approaches: recent trends in designing novel therapeutics for amyloidogenesis. J Mater Chem B 2025; 13:858-881. [PMID: 39664012 DOI: 10.1039/d4tb01890g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Amyloidosis is a condition marked by misfolded proteins that build up in tissues and eventually destroy organs. It has been connected to a number of fatal illnesses, including non-neuropathic and neurodegenerative conditions, which in turn have a significant influence on the worldwide health sector. The inability to identify the underlying etiology of amyloidosis has hampered efforts to find a treatment for the condition. Despite the identification of a multitude of putative pathogenic variables that may operate independently or in combination, the molecular mechanisms responsible for the development and progression of the disease remain unclear. A thorough investigation into protein aggregation and the impacts of toxic aggregated species will help to clarify the cytotoxicity of aggregation-mediated cellular apoptosis and lay the groundwork for future studies aimed at creating effective treatments and medications. This review article provides a thorough summary of the combination of various experimental and computational approaches to modulate amyloid aggregation. Further, an overview of the latest developments of novel therapeutic agents is given, along with a discussion of the possible obstacles and viewpoints on this developing field. We believe that the information provided by this review will help scientists create innovative treatment strategies that affect the way proteins aggregate.
Collapse
Affiliation(s)
- Pooja Ghosh
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies & Research (JISIASR) Kolkata, JIS University, GP Block, Sector-5, Salt Lake, Kolkata 700091, West Bengal, India.
| | - Agnibin Kundu
- Department of Medicine, District Hospital Howrah, 10, Biplabi Haren Ghosh Sarani Lane, Howrah 711101, West Bengal, India
| | - Debabani Ganguly
- Centre for Health Science & Technology, JIS Institute of Advanced Studies & Research (JISIASR) Kolkata, JIS University, GP Block, Sector-5, Salt Lake, Kolkata 700091, West Bengal, India.
| |
Collapse
|
5
|
İrişli S, Çakır A, Günnaz S. Synthesis of novel pyrimidine-based Schiff base complexes: Targeting Amyloid-β aggregation in Alzheimer's disease. Bioorg Chem 2025; 154:107999. [PMID: 39637485 DOI: 10.1016/j.bioorg.2024.107999] [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: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
A novel Schiff base with imine/amine donors, 5-((3,3-diphenylalilidene)amino)pyrimidine-4-amine (L), and its new Platinum(II) and Ruthenium(II) complexes (I and II) were synthesized and characterized using FT-IR, 1H NMR, 13C NMR, mass spectrometry and elemental analyses. The ability of these complexes to inhibit amyloid beta (Aβ1-42) aggregation was evaluated using the human neuroblastoma cell line (SH-SY5Y). The complexes effectively inhibited Aβ1-42 aggregation at a 1:1 M ratio. Both complexes increased cell viability up to 80 % at concentrations of 10 μM. At this concentration, the cell viability value found by Aβ1-42 aggregation is around 65 %. Aggregation kinetics were fluorometrically monitored using Thioflavin T. These findings were further supported by scanning electron microscopy and transmission electron microscopy. In addition, the interaction of the complexes with Aβ1-16 was investigated using MALDI-TOF/MS and 1H NMR spectroscopy. All findings showed that Aβ1-42 in both complexes is active in the inhibition of amyloid aggregation.
Collapse
Affiliation(s)
- Sevil İrişli
- Department of Chemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| | - Aslı Çakır
- Department of Chemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| | - Salih Günnaz
- Department of Chemistry, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey.
| |
Collapse
|
6
|
Nabi F, Ahmad O, Fatima A, Ahmad A, Sharma J, Khan RH. Small molecule inhibits BACE1 activity by a dual mechanism confirmed by simulations-based study. J Biomol Struct Dyn 2024:1-13. [PMID: 39633599 DOI: 10.1080/07391102.2024.2435641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/29/2024] [Indexed: 12/07/2024]
Abstract
Alzheimer's disease (AD) is a progressive and largely incurable neurodegenerative disorder that affects millions of people worldwide. It is characterised by the accumulation of amyloid-beta plaques and neurofibrillary tangles in the brain. It is commenced by cleavage of amyloid precursor protein (APP) by β-secretase, β-site amyloid precursor protein cleaving enzyme (BACE1; also called Asp2, memapsin 2). Therefore, BACE1 is a prime target for developing therapeutics against AD. In this study, we have identified a small molecule that potentially inhibits the activity of BACE1 by interacting with the active site residues. Also, the flap region seems to be involved in enhancing the stability of the small molecule at the active site. We have used Umibecestat (CNP-520) as a positive control. Our in silico results show that the identified molecule has a much better orientation at the active site of BACE1 than Umibecestat and inhibits by blocking the active site and modulating flap dynamics. We have utilised virtual high-throughput screening assay, ADME profiling, and blood-brain-barrier crossing ability to narrow down potential leads. The two shortlisted molecules were then subjected to atomistic molecular dynamics simulations study. Overall, our study proposes a much better inhibitor and a rational molecule for lead development against AD.
Collapse
Affiliation(s)
- Faisal Nabi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Owais Ahmad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Aiman Fatima
- Department of Botany, Aligarh Muslim University, Aligarh, India
| | - Aamna Ahmad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
- Integral University, Lucknow, India
| | - Jyoti Sharma
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
7
|
Navale GR, Ahmed I, Lim MH, Ghosh K. Transition Metal Complexes as Therapeutics: A New Frontier in Combatting Neurodegenerative Disorders through Protein Aggregation Modulation. Adv Healthc Mater 2024; 13:e2401991. [PMID: 39221545 DOI: 10.1002/adhm.202401991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Neurodegenerative disorders (NDDs) are a class of debilitating diseases that progressively impair the protein structure and result in neurological dysfunction in the nervous system. Among these disorders, Alzheimer's disease (AD), prion diseases such as Creutzfeldt-Jakob disease (CJD), and Parkinson's disease (PD) are caused by protein misfolding and aggregation at the cellular level. In recent years, transition metal complexes have gained significant attention for their potential applications in diagnosing, imaging, and curing these NDDs. These complexes have intriguing possibilities as therapeutics due to their diverse ligand systems and chemical properties and can interact with biological systems with minimal detrimental effects. This review focuses on the recent progress in transition metal therapeutics as a new era of hope in the battle against AD, CJD, and PD by modulating protein aggregation in vitro and in vivo. It may shed revolutionary insights into unlocking new opportunities for researchers to develop metal-based drugs to combat NDDs.
Collapse
Affiliation(s)
- Govinda R Navale
- Department of Chemistry, Indian Institute of Technology, Roorkee, 247667, India
| | - Imtiaz Ahmed
- Department of Chemistry, Indian Institute of Technology, Roorkee, 247667, India
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kaushik Ghosh
- Department of Chemistry, Indian Institute of Technology, Roorkee, 247667, India
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, 247667, India
| |
Collapse
|
8
|
Nabi F, Ahmad O, Khan A, Hassan MN, Hisamuddin M, Malik S, Chaari A, Khan RH. Natural compound plumbagin based inhibition of hIAPP revealed by Markov state models based on MD data along with experimental validations. Proteins 2024; 92:1070-1084. [PMID: 38497314 DOI: 10.1002/prot.26682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024]
Abstract
Human islet amyloid polypeptide (amylin or hIAPP) is a 37 residue hormone co-secreted with insulin from β cells of the pancreas. In patients suffering from type-2 diabetes, amylin self-assembles into amyloid fibrils, ultimately leading to the death of the pancreatic cells. However, a research gap exists in preventing and treating such amyloidosis. Plumbagin, a natural compound, has previously been demonstrated to have inhibitory potential against insulin amyloidosis. Our investigation unveils collapsible regions within hIAPP that, upon collapse, facilitates hydrophobic and pi-pi interactions, ultimately leading to aggregation. Intriguingly plumbagin exhibits the ability to bind these specific collapsible regions, thereby impeding the aforementioned interactions that would otherwise drive hIAPP aggregation. We have used atomistic molecular dynamics approach to determine secondary structural changes. MSM shows metastable states forming native like hIAPP structure in presence of PGN. Our in silico results concur with in vitro results. The ThT assay revealed a striking 50% decrease in fluorescence intensity at a 1:1 ratio of hIAPP to Plumbagin. This finding suggests a significant inhibition of amyloid fibril formation by plumbagin, as ThT fluorescence directly correlates with the presence of these fibrils. Further TEM images revealed disappearance of hIAPP fibrils in plumbagin pre-treated hIAPP samples. Also, we have shown that plumbagin disrupts the intermolecular hydrogen bonding in hIAPP fibrils leading to an increase in the average beta strand spacing, thereby causing disaggregation of pre-formed fibrils demonstrating overall disruption of the aggregation machinery of hIAPP. Our work is the first to report a detailed atomistic simulation of 22 μs for hIAPP. Overall, our studies put plumbagin as a potential candidate for both preventive and therapeutic candidate for hIAPP amyloidosis.
Collapse
Affiliation(s)
- Faisal Nabi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Owais Ahmad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Adeeba Khan
- Zakir Hussain College of Engineering and Technology, Aligarh Muslim University, Aligarh, India
| | - Md Nadir Hassan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Malik Hisamuddin
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Sadia Malik
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Ali Chaari
- Premedical Division, Weill Cornell Medicine Qatar, Qatar Foundation, Doha, Qatar
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
9
|
Zakaria N, Wan Harun WMRS, Mohammad Latif MA, Azaman SNA, Abdul Rahman MB, Faujan NH. Effects of anthocyanidins on the conformational transition of Aβ (1-42) peptide: Insights from molecular docking and molecular dynamics simulations. J Mol Graph Model 2024; 129:108732. [PMID: 38412813 DOI: 10.1016/j.jmgm.2024.108732] [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: 10/20/2023] [Revised: 12/31/2023] [Accepted: 02/14/2024] [Indexed: 02/29/2024]
Abstract
Recent evidence from in vitro and in vivo studies has shown that anthocyanins and anthocyanidins can reduce and inhibit the amyloid beta (Aβ) species, one of the hallmarks of Alzheimer's disease (AD). However, their inhibition mechanisms on Aβ species at molecular details remain elusive. Therefore, in the present study, molecular modelling methods were employed to investigate their inhibitory mechanisms on Aβ(1-42) peptide. The results highlighted that anthocyanidins effectively inhibited the conformational transitions of helices into beta-sheet (β-sheet) conformation within Aβ(1-42) peptide by two different mechanisms: 1) the obstruction of two terminals from coming into contact due to the binding of anthocyanidins with residues of N- and second hydrophobic core (SHC)-C-terminals, and 2) the prevention of the folding process due to the binding of anthocyanidin with the central polar (Asp23 and Lys28) and native helix (Asp23, Lys28, and Leu34) residues. These new findings on the inhibition of β-sheet formation by targeting both N- and SHC-C-terminals, and the long-established target, D23-K28 salt bridge residues, not with the conventional central hydrophobic core (CHC) as reported in the literature, might aid in designing more potent inhibitors for AD treatment.
Collapse
Affiliation(s)
- Norzalina Zakaria
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Macromolecular Simulation Laboratory, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Integrated Chemical Biophysics Research, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | | | - Muhammad Alif Mohammad Latif
- Center of Foundation Studies for Agricultural Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Macromolecular Simulation Laboratory, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Integrated Chemical Biophysics Research, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Siti Nor Ani Azaman
- Center of Foundation Studies for Agricultural Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohd Basyaruddin Abdul Rahman
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Macromolecular Simulation Laboratory, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Integrated Chemical Biophysics Research, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Nur Hana Faujan
- Center of Foundation Studies for Agricultural Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Macromolecular Simulation Laboratory, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Natural Medicine and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
10
|
Grcic L, Leech G, Kwan K, Storr T. Targeting misfolding and aggregation of the amyloid-β peptide and mutant p53 protein using multifunctional molecules. Chem Commun (Camb) 2024; 60:1372-1388. [PMID: 38204416 DOI: 10.1039/d3cc05834d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Biomolecule misfolding and aggregation play a major role in human disease, spanning from neurodegeneration to cancer. Inhibition of these processes is of considerable interest, and due to the multifactorial nature of these diseases, the development of drugs that act on multiple pathways simultaneously is a promising approach. This Feature Article focuses on the development of multifunctional molecules designed to inhibit the misfolding and aggregation of the amyloid-β (Aβ) peptide in Alzheimer's disease (AD), and the mutant p53 protein in cancer. While for the former, the goal is to accelerate the removal of the Aβ peptide and associated aggregates, for the latter, the goal is reactivation via stabilization of the active folded form of mutant p53 protein and/or aggregation inhibition. Due to the similar aggregation pathway of the Aβ peptide and mutant p53 protein, a common therapeutic approach may be applicable.
Collapse
Affiliation(s)
- Lauryn Grcic
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| | - Grace Leech
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| | - Kalvin Kwan
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| | - Tim Storr
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| |
Collapse
|
11
|
Hussain R, Rubab SL, Maryam A, Ashraf T, Arshad M, Lal K, Sumrra SH, Ashraf S, Ali B. Synthesis, Spectroscopic and Nonlinear Optical Properties, and Antimicrobial Activity of Cu(II), Co(II), and Ni(II) Complexes: Experimental and Theoretical Studies. ACS OMEGA 2023; 8:42598-42609. [PMID: 38024690 PMCID: PMC10652729 DOI: 10.1021/acsomega.3c05322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023]
Abstract
Currently, we report the preparation of transition metal complexes Co(II), Ni(II), and Cu(II) of hydrazone Schiff base ligands, which are obtained by the condensation reaction of substituted salicylaldehyde and hydrazines. The synthesized hydrazone ligands and their metal complexes were characterized by spectroscopic methods such as Fourier transform infrared (FT-IR), UV-vis, nuclear magnetic resonance (1H NMR and C13 NMR), and mass spectrometry analyses. All of the quantum chemistry calculations were performed using DFT executed in the Gaussian 09 software package. The geometry was optimized by using the density functional theory (DFT) approximation at the B3LYP level with a basis set of 6-31G (d, p). There was excellent agreement between the FT-IR values obtained experimentally and those obtained theoretically for the test compounds. It is worth noting that none of the optimized geometries for any of the Schiff base and metal complexes had any eigenvalues that were negative, indicating that these geometries represent the true minimum feasible energy surfaces. We also analyzed the electrostatic potential of the molecule and NBO calculation at the same level of theory. Gauss View 6 was utilized for the file organization of the input data. Gauss View 6.0, Avogadro, and Chemcraft were used to determine the data. Additionally, synthesized compounds were screened for antimicrobial activity against Gram-negative bacteria (Salmonella typhi, Escherichia coli) and Gram-positive bacteria (Bacillus halodurans, Micrococcus luteus) and two fungal strains (Aspergillus flavus, Aspergillus niger). These research findings have established the potential of ligands and their metal complexes as antimicrobial agents. Additionally, the compounds demonstrated promising nonlinear optical (NLO) properties, with potential applications across a wide range of contemporary technologies.
Collapse
Affiliation(s)
- Riaz Hussain
- Department
of Chemistry, The Education University Lahore
D.G Khan campus, Dera Ghazi Khan32200,Pakistan
| | - Syed Laila Rubab
- Department
of Chemistry, The Education University Lahore
D.G Khan campus, Dera Ghazi Khan32200,Pakistan
| | - Afifa Maryam
- Institute
of Chemistry, Khwaja Fareed University of Engineering & Information
Technology, Rahim
Yar Khan 64200, Pakistan
| | - Tuba Ashraf
- Institute
of Chemistry, Khwaja Fareed University of Engineering & Information
Technology, Rahim
Yar Khan 64200, Pakistan
| | - Muhammad Arshad
- Department
of Chemical Engineering, College of Engineering, King Khalid University, Abha 62529, Saudi Arabia
| | - Kiran Lal
- Department
of Chemistry, The Women University Multan, Multan 60000, Pakistan
| | - Sajjad H. Sumrra
- Department
of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | - Shafaq Ashraf
- Institute
of Chemistry, Khwaja Fareed University of Engineering & Information
Technology, Rahim
Yar Khan 64200, Pakistan
| | - Bakhat Ali
- Institute
of Chemistry, Khwaja Fareed University of Engineering & Information
Technology, Rahim
Yar Khan 64200, Pakistan
| |
Collapse
|
12
|
Wang M, Zhang Z, Jing B, Dong X, Guo K, Deng J, Wang Z, Wan W, Jin W, Gao Z, Liu Y. Tailoring the Amphiphilicity of Fluorescent Protein Chromophores to Detect Intracellular Proteome Aggregation in Diverse Biological Samples. Anal Chem 2023; 95:11751-11760. [PMID: 37506028 DOI: 10.1021/acs.analchem.3c01903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
The formation of amorphous misfolded and aggregated proteins is a hallmark of proteome stress in diseased cells. Given its lack of defined targeting sites, the rational design of intracellular proteome aggregation sensors has been challenging. Herein, we modulate the amphiphilicity of fluorescent protein chromophores to enable selective detection of aggregated proteins in different biological samples, including recombinant proteins, stressed live cells, intoxicated mouse liver tissue, and human hepatocellular carcinoma tissue. By tuning the number of hydroxyl groups, we optimize the selectivity of fluorescent protein chromophores toward aggregated proteins in these biological samples. In recombinant protein applications, the most hydrophobic P0 (cLogP = 5.28) offers the highest fold change (FC = 31.6), sensitivity (LLOD = 0.1 μM), and brightness (Φ = 0.20) upon binding to aggregated proteins. In contrast, P4 of balanced amphiphilicity (cLogP = 2.32) is required for selective detection of proteome stresses in live cells. In mouse and human liver histology tissues, hydrophobic P1 exhibits the best performance in staining the aggregated proteome. Overall, the amphiphilicity of fluorescent chromophores governs the sensor's performance by matching the diverse nature of different biological samples. Together with common extracellular amyloid sensors (e.g., Thioflavin T), these sensors developed herein for intracellular amorphous aggregation complement the toolbox to study protein aggregation.
Collapse
Affiliation(s)
- Mengdie Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenduo Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Biao Jing
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Kun Guo
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Jintai Deng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Zhiming Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Zhenming Gao
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| |
Collapse
|
13
|
Singh S, Navale GR, Agrawal S, Singh HK, Singla L, Sarkar D, Sarma M, Choudhury AR, Ghosh K. Design and synthesis of ruthenium complexes and their studies on the inhibition of amyloid β (1-42) peptide aggregation. Int J Biol Macromol 2023; 239:124197. [PMID: 36972817 DOI: 10.1016/j.ijbiomac.2023.124197] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
Misfolding and protein aggregation have been linked to numerous human neurodegenerative disorders such as Alzheimer's, prions, and Parkinson's. Due to their interesting photophysical properties, ruthenium (Ru) complexes have received considerable attention in studying protein aggregation. In this study, we synthesized the novel Ru complexes ([Ru(p-cymene)Cl(L-1)][PF6](Ru-1), and [Ru(p-cymene)Cl(L-2)][PF6](Ru-2)) and investigated their inhibitory activity against the bovine serum albumin (BSA) aggregation and the Aβ1-42 peptides amyloid formation. Several spectroscopic methods were used to characterize the complexes, and the molecular structure was determined by X-ray crystallography. Amyloid aggregation and inhibition activity were examined using the Thioflavin-T (ThT) assay, and secondary structures were analyzed by circular dichroism (CD) spectroscopy and transmission electron microscopy (TEM). The cell viability assay was carried out on the neuroblastoma cell line, revealing that the Ru-2 complex showed better protective effects against Aβ1-42 peptide toxicity on neuro-2a cells than the Ru-1 complex. Molecular docking studies elucidate binding sites and interactions between the Ru-complexes and the Aβ1-42 fibrils. The experimental studies revealed that these complexes significantly inhibited BSA aggregation and Aβ1-42 amyloid fibril formation at 1:3 and 1:1 equimolar concentrations, respectively. Antioxidant assays demonstrated that these complexes act as antioxidants, protecting from amyloid-induced oxidative stress. Molecular docking studies with the monomeric Aβ1-42 (PDB: 1IYT) show hydrophobic interaction, and both complexes bind preferably in the central region of the peptide and coordinate with two binding sites of the peptide. Hence, we suggest that the Ru-based complexes could be applied as a potential agent in metallopharmaceutical research against Alzheimer's disease.
Collapse
Affiliation(s)
- Sain Singh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Govinda R Navale
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Sonia Agrawal
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Haobam Kisan Singh
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Labhini Singla
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Dhiman Sarkar
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Manabendra Sarma
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Anghuman Roy Choudhury
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Kaushik Ghosh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India; Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, India.
| |
Collapse
|
14
|
Li Q, Wen J, Yan Z, Sun H, Song E, Song Y. Mechanistic Insights of TiO 2 Nanoparticles with Different Surface Charges on Aβ 42 Peptide Early Aggregation: An In Vitro and In Silico Study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1997-2007. [PMID: 36706054 DOI: 10.1021/acs.langmuir.2c03065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Humans may intendedly or unintendedly be exposed to nanomaterials through food, water, and air. Upon exposure, nanomaterials can pierce the bloodstream and translocate to secondary organs, including the brain, which warrants increased concern for the potential health impacts of nanomaterials. Due to their large surface area and interaction energy, nanomaterials can adsorb surrounding proteins. The misfolding and self-aggregation of amyloid-β (Aβ) have been considered significant factors in the pathogenesis of Alzheimer's disease. We thus hypothesize that brain-targeted nanomaterials may modulate Aβ aggregation and cause related neurotoxicity. Here, we showed that TiO2 nanoparticles (NPs) and their aminated analogue (TiO2-NH2 NPs) adsorb the Aβ42 peptide and accelerate its early oligomerization. Molecular dynamics simulation indicated that the adsorption onto TiO2 NPs and TiO2-NH2 NPs surfaces can stabilize the β-sheet-rich conformations formed by the Aβ42 peptide. The binding sites between TiO2-NH2 NPs and the Aβ42 oligomer surface were mainly concentrated in the hydrophobic core region, and the β-sheet conformation spontaneously formed by Aβ42 oligomers can be better stabilized through a hydrogen bond, electrostatic attraction, and hydrophobic interaction. This study will further help in the understanding of nanomaterial-related neurotoxicities and the regulation of their applications.
Collapse
Affiliation(s)
- Qiong Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Jing Wen
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
| | - Ziyi Yan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
| | - Hang Sun
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
| |
Collapse
|
15
|
Kwak J, Woo J, Park S, Lim MH. Rational design of photoactivatable metal complexes to target and modulate amyloid-β peptides. J Inorg Biochem 2023; 238:112053. [PMID: 36347209 DOI: 10.1016/j.jinorgbio.2022.112053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/25/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022]
Abstract
The accumulation of amyloid-β (Aβ) aggregates is found in the brains of Alzheimer's disease patients. Thus, numerous efforts have been made to develop chemical reagents capable of targeting Aβ peptides and controlling their aggregation. In particular, tunable coordination and photophysical properties of transition metal complexes, with variable oxidation and spin states on the metal centers, can be utilized to probe Aβ aggregates and alter their aggregation profiles. In this review, we illustrate some rational strategies for designing photoactivatable metal complexes as chemical sensors for Aβ peptides or modulators against their aggregation pathways, with some examples.
Collapse
Affiliation(s)
- Jimin Kwak
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Junhyeok Woo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seongmin Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
16
|
Tajahmadi S, Molavi H, Ahmadijokani F, Shamloo A, Shojaei A, Sharifzadeh M, Rezakazemi M, Fatehizadeh A, Aminabhavi TM, Arjmand M. Metal-organic frameworks: A promising option for the diagnosis and treatment of Alzheimer's disease. J Control Release 2023; 353:1-29. [PMID: 36343762 DOI: 10.1016/j.jconrel.2022.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/22/2022]
Abstract
Beta-amyloid (Aβ) peptide is one of the main characteristic biomarkers of Alzheimer's disease (AD). Previous clinical investigations have proposed that unusual concentrations of this biomarker in cerebrospinal fluid, blood, and brain tissue are closely associated with the AD progression. Therefore, the critical point of early diagnosis, prevention, and treatment of AD is to monitor the levels of Aβ. In view of the potential of metal-organic frameworks (MOFs) for diagnosing and treating the AD, much attention has been focused in recent years. This review discusses the latest advances in the applications of MOFs for the early diagnosis of AD via fluorescence and electrochemiluminescence (ECL) detection of AD biomarkers, fluorescence detection of the main metal ions in the brain (Zn2+, Cu2+, Mn2+, Fe3+, and Al3+) in addition to magnetic resonance imaging (MRI) of the Aβ plaques. The current challenges and future strategies for translating the in vitro applications of MOFs into in vivo diagnosis of the AD are discussed.
Collapse
Affiliation(s)
- Shima Tajahmadi
- Institute for Nanoscience and Nanotechnology (INST), Sharif University of Technology, Tehran, Iran
| | - Hossein Molavi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran; Department of Chemistry, Institute for Advanced Studies in Basic Science (IASBS), Gava Zang, Zanjan 45137-66731, Iran
| | - Farhad Ahmadijokani
- Nanomaterials and Polymer Nanocomposites Laboratory, School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Amir Shamloo
- Institute for Nanoscience and Nanotechnology (INST), Sharif University of Technology, Tehran, Iran; Department of Mechanical Engineering, Sharif University of Technology, Azadi Ave., Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran 11155-9161, Iran.
| | - Akbar Shojaei
- Institute for Nanoscience and Nanotechnology (INST), Sharif University of Technology, Tehran, Iran; Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mashallah Rezakazemi
- Faculty of Chemical and Materials Engineering, Shahrood University of Technology, Shahrood, Iran
| | - Ali Fatehizadeh
- Department of Environmental Health Engineering, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran; Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tejraj M Aminabhavi
- School of Advanced Sciences, KLE Technological University, Hubballi, Karnataka 580 031, India; School of Engineering, UPES, Bidholi, Dehradun, Uttarakhand 248 007, India.
| | - Mohammad Arjmand
- Nanomaterials and Polymer Nanocomposites Laboratory, School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada.
| |
Collapse
|
17
|
Bassan GA, Marchesan S. Peptide-Based Materials That Exploit Metal Coordination. Int J Mol Sci 2022; 24:ijms24010456. [PMID: 36613898 PMCID: PMC9820281 DOI: 10.3390/ijms24010456] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Metal-ion coordination has been widely exploited to control the supramolecular behavior of a variety of building blocks into functional materials. In particular, peptides offer great chemical diversity for metal-binding modes, combined with inherent biocompatibility and biodegradability that make them attractive especially for medicine, sensing, and environmental remediation. The focus of this review is the last 5 years' progress in this exciting field to conclude with an overview of the future directions that this research area is currently undertaking.
Collapse
|
18
|
Shen D, Jin W, Zhao Q, Wang M, Zhang B, Feng H, Wan W, Bai Y, Lyu H, Sun J, Zhang L, Liu Y. Covalent Solvatochromic Proteome Stress Sensor Based on the Schiff Base Reaction. Anal Chem 2022; 94:14143-14150. [PMID: 36194526 DOI: 10.1021/acs.analchem.2c01281] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Covalent-type probes or sensors have been seldom reported for aggregated proteins. Herein, we reported a series of covalent solvatochromic probes to selectively modify and detect aggregated proteomes through the Schiff base reaction. Such covalent modification was discovered by serendipity using the P1 probe with an aldehyde functional group, exhibiting enhanced fluorescence intensity and unusually large blue shift upon protein aggregation. Supported by the biochemical and mass spectrometry results, we identified that this probe can modify the lysine residue of aggregated proteins selectively over folded ones via the Schiff base reaction. The generality of designing such a covalent-type probe was demonstrated in multiple probe scaffolds using different model proteins. Finally, we exploited the distinct solvatochromism of P1 after Schiff base linkage with aggregated proteins to visualize the distinct morphology of aggregated proteomes, as well as to quantify the polarity heterogeneity inside it. This work may intrigue the exploration of other chemical reaction types to covalently functionalize aggregated proteins that were difficult to analyze.
Collapse
Affiliation(s)
- Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Qun Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Mengdie Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.,University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Beirong Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.,University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Feng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.,University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Yulong Bai
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.,University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Haochen Lyu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Jialu Sun
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| |
Collapse
|
19
|
Schiff base nanoarchitectonics for supramolecular assembly of dipeptide as drug carriers. J Colloid Interface Sci 2022; 630:161-169. [DOI: 10.1016/j.jcis.2022.09.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022]
|
20
|
Yang H, Li J, Li X, Ma L, Hou M, Zhou H, Zhou R. Based on molecular structures: Amyloid-β generation, clearance, toxicity and therapeutic strategies. Front Mol Neurosci 2022; 15:927530. [PMID: 36117918 PMCID: PMC9470852 DOI: 10.3389/fnmol.2022.927530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Amyloid-β (Aβ) has long been considered as one of the most important pathogenic factors in Alzheimer’s disease (AD), but the specific pathogenic mechanism of Aβ is still not completely understood. In recent years, the development of structural biology technology has led to new understandings about Aβ molecular structures, Aβ generation and clearance from the brain and peripheral tissues, and its pathological toxicity. The purpose of the review is to discuss Aβ metabolism and toxicity, and the therapeutic strategy of AD based on the latest progress in molecular structures of Aβ. The Aβ structure at the atomic level has been analyzed, which provides a new and refined perspective to comprehend the role of Aβ in AD and to formulate therapeutic strategies of AD.
Collapse
Affiliation(s)
- Hai Yang
- Department of Neurology, Army Medical Center of PLA, Chongqing, China
| | - Jinping Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaoxiong Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Linqiu Ma
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mingliang Hou
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huadong Zhou
- Department of Neurology, Army Medical Center of PLA, Chongqing, China
| | - Rui Zhou
- Southwest Hospital, Army Medical University, Chongqing, China
- *Correspondence: Rui Zhou,
| |
Collapse
|
21
|
Brue CR, Dukes MW, Masotti M, Holmgren R, Meade TJ. Functional Disruption of Gli1-DNA Recognition via a Cobalt(III) Complex. ChemMedChem 2022; 17:e202200025. [PMID: 35302712 PMCID: PMC10826845 DOI: 10.1002/cmdc.202200025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Indexed: 12/29/2022]
Abstract
The aberrant activation of the Gli family of zinc finger transcription factors (ZFTFs) is associated with several types of human cancer, including medulloblastoma and basal cell carcinoma. We have reported the use of cobalt(III) Schiff-base complexes (Co(III)-sb) as potent inhibitors of ZFTFs in vivo. These complexes inhibit transcription by displacing the zinc finger domain's structural Zn(II) ion, destabilizing the alpha helix necessary for DNA recognition. Here, we describe the use of Co(III)-sb complexes for the selective inhibition of Gli1. Spectroscopic and computational studies of the Gli1 DNA binding domain found that Co(III)-sb displaced Zn(II) through direct coordination with the His residues of the Cys2 His2 Zn(II) binding site. As a result, there is a dose-dependent degradation of the alpha-helix content in the DNA binding domain of Gli1 and corresponding inhibition of consensus sequence recognition. We conclude that this strategy is well suited for the development of new and potent inhibitors of Gli1.
Collapse
Affiliation(s)
- Christopher R Brue
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Meghan W Dukes
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Meghan Masotti
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Robert Holmgren
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| | - Thomas J Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL, 60208-3113, USA
| |
Collapse
|
22
|
Xu S, Sun Y, Dong X. Design of Gallic Acid-Glutamine Conjugate and Chemical Implications for Its Potency Against Alzheimer's Amyloid-β Fibrillogenesis. Bioconjug Chem 2022; 33:677-690. [PMID: 35380783 DOI: 10.1021/acs.bioconjchem.2c00073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Epigallocatechin-3-gallate (EGCG) has been widely recognized as a potent inhibitor of Alzheimer's amyloid-β (Aβ) fibrillogenesis. We found that gallic acid (GA) has superior inhibitory effects over EGCG at the same mass concentrations and assumed the pivotal role of the carboxyl group in GA. Therefore, we designed five GA-derivatives to investigate the significance of carboxyl groups in modulating Aβ fibrillogenesis, including carboxyl-amidated GA (GA-NH2), GA-glutamic acid conjugate (GA-E), and GA-E derivatives with amidated either of the two carboxyl groups (GA-Q and GA-E-NH2) or with two amidated-carboxyl groups (GA-Q-NH2). Intriguingly, only GA-Q shows significantly stronger potency than GA and extends the life span of the AD transgenic nematode by over 30%. Thermodynamic studies reveal that GA-Q has a strong binding affinity for Aβ42 with two binding sites, one stronger (site 1, Ka1 = 3.1 × 106 M-1) and the other weaker (site 2, Ka2 = 0.8 × 106 M-1). In site 1, hydrogen bonding, electrostatic interactions, and hydrophobic interactions all have contributions, while in site 2, only hydrogen bonding and electrostatic interactions work. The two sites are confirmed by molecular simulations, and the computations specified the key residues. GA-Q has strong binding to Asp23, Gly33, Gly38, Ala30, Ile31, and Leu34 via hydrogen bonding and electrostatic interactions, while it interacts with Phe19, Ala21 Gly25, and Asn27 via hydrophobic interactions. Consequently, GA-Q destroys Asp23-Lys28 salt bridges and restricts β-sheet/bridge structures. The thermodynamic and molecular insight into the GA-Q functions on inhibiting Aβ fibrillogenesis would pave a new way to the design of potent molecules against Alzheimer's amyloid.
Collapse
Affiliation(s)
- Shaoying Xu
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| |
Collapse
|
23
|
Zhang Z, Cao Y, Yuan Q, Liu C, Duan X, Tang Y. Multifunctional fluorescent probe for effective visualization, inhibition, and detoxification of β-amyloid aggregation via covalent binding. Chem Commun (Camb) 2022; 58:3957-3960. [PMID: 35244642 DOI: 10.1039/d2cc00318j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A multifunctional reactive fluorescent probe DTB was constructed for biosensing, aggregation inhibition, and toxicity alleviation of β-amyloid. The synergistic effect of hydrophobic interaction and covalent interaction makes DTB have more stable binding and better selectivity to Aβ. The detoxification effect of DTB on Aβ aggregates was also verified in live nerve cells and microglia cells. Furthermore, DTB exhibits an excellent staining of Aβ plaques.
Collapse
Affiliation(s)
- Ziqi Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, P. R. China.
| | - Yue Cao
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, P. R. China.
| | - Qiong Yuan
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, P. R. China.
| | - Chenghui Liu
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, P. R. China.
| | - Xinrui Duan
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, P. R. China.
| | - Yanli Tang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, P. R. China.
| |
Collapse
|
24
|
Meiss CJ, Bothwell PJ, Webb MI. Ruthenium(II)–arene complexes with chelating quinoline ligands as anti-amyloid agents. CAN J CHEM 2022. [DOI: 10.1139/cjc-2021-0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent recognition of the soluble form of the amyloid-beta (Aβ) peptide as a neurotoxic agent in Alzheimer’s disease (AD) has spurred the development of agents to target this species. Because Aβ is known to chelate metal ions in solution, metal-based therapeutics are uniquely suited to exploit this affinity, where coordination to Aβ has been shown to impact the neurotoxicity of the peptide. Ruthenium(II)–arene complexes are unique candidates for evaluation, as one face of the molecule is blocked by the hydrophobic arene ring, while coordination to the Aβ peptide can occur on the other side of the molecule. We have prepared and evaluated two Ru(II)–arene complexes with chelating quinoline-based ligands, Ru1 and Ru2, for their respective anti-amyloid abilities. Although both complexes decreased the aggregation of soluble Aβ, Ru1 displayed promise in disrupting formed aggregates of the peptide. These findings represent an exciting new avenue for therapeutic development in AD, where both sides of the aggregation equilibrium are affected.
Collapse
Affiliation(s)
- Cade J. Meiss
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, USA
| | - Paige J. Bothwell
- Core Microscope Facility, Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115, USA
| | - Michael I. Webb
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, USA
| |
Collapse
|
25
|
Dong X, Wan W, Zeng L, Jin W, Huang Y, Shen D, Bai Y, Zhao Q, Zhang L, Liu Y, Gao Z. Regulation of Fluorescence Solvatochromism To Resolve Cellular Polarity upon Protein Aggregation. Anal Chem 2021; 93:16447-16455. [PMID: 34859995 DOI: 10.1021/acs.analchem.1c03401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Common solvatochromic fluorophores exhibit a bathochromic fluorescence emission wavelength shift accompanied by intensity attenuation due to the presence of nonradiative decay pathways at the excited state. Such intrinsic but inevitable fluorescence quenching of solvatochromism impedes its applications to faithfully quantify local polarity, especially in a polar environment. Herein, we report a new donor-π-acceptor (D-π-A) type solvatochromic fluorophore scaffold containing a perfluorophenyl group that exhibits both a solvatochromic emission wavelength shift and a controllable emission intensity upon polarity fluctuation. The regulation of fluorescence solvatochromism and colors was achieved by tuning the aryl donors. We exploited such desired solvatochromism of these probes to monitor protein misfolding and aggregation via wavelength shift. Finally, the polarity of pathogenic aggregated proteins was quantified by HaloTag bioorthogonal labeling technology in live cells. While much effort has been devoted to resolving the morphology of pathogenic aggregated proteins, this work provides quantitative hints regarding the chemical information at this disease-related protein interphase.
Collapse
Affiliation(s)
- Xuepeng Dong
- The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, P. R. China
| | - Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Lianggang Zeng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Yanan Huang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Yulong Bai
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China.,University of the Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Qun Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Zhenming Gao
- The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, P. R. China
| |
Collapse
|
26
|
Giriraj K, Mohamed Kasim MS, Balasubramaniam K, Thangavel SK, Venkatesan J, Suresh S, Shanmugam P, Karri C. Various coordination modes of new coumarin Schiff bases toward Cobalt (III) ion: Synthesis, spectral characterization, in vitro cytotoxic activity, and investigation of apoptosis. Appl Organomet Chem 2021. [DOI: 10.1002/aoc.6536] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Kalaiarasi Giriraj
- Department of Chemistry Karpagam Academy of Higher Education (Deemed to be University) Coimbatore 641021 India
| | - Mohamed Subarkhan Mohamed Kasim
- The First Affiliated Hospital; Key Laboratory of Combined Multi Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou PR China
| | - Keerthana Balasubramaniam
- Department of Microbiology Karpagam Academy of Higher Education (Deemed to be University) Coimbatore 641021 India
| | - Sathiya Kamatchi Thangavel
- Centre for Organometallic Chemistry, School of Chemistry Bharathidasan University Tiruchirappalli 620024 India
| | - Janani Venkatesan
- Department of Chemistry Karpagam Academy of Higher Education (Deemed to be University) Coimbatore 641021 India
| | - Sharmila Suresh
- Department of Chemistry Karpagam Academy of Higher Education (Deemed to be University) Coimbatore 641021 India
| | - Pritha Shanmugam
- Department of Chemistry Karpagam Academy of Higher Education (Deemed to be University) Coimbatore 641021 India
| | - Chiranjeevi Karri
- Department of Chemistry Karpagam Academy of Higher Education (Deemed to be University) Coimbatore 641021 India
| |
Collapse
|
27
|
Gong Z, Zhou B, Liu X, Cao J, Hong Z, Wang J, Sun X, Yuan X, Tan H, Ji H, Bai J. Enzyme-Induced Transformable Peptide Nanocarriers with Enhanced Drug Permeability and Retention to Improve Tumor Nanotherapy Efficacy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:55913-55927. [PMID: 34784165 DOI: 10.1021/acsami.1c17917] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Temporal persistence is as important for nanocarriers as spatial accuracy. However, because of the insufficient aggreagtion and short retention time of chemotherapy drugs in tumors, their clinical application is greatly limited. A drug delivery approach dependent on the sensitivity to an enzyme present in the microenvironment of the tumor is designed to exhibit different sizes in different sites, achieving enhanced drug permeability and retention to improve tumor nanotherapy efficacy. In this work, we report a small-molecule peptide drug delivery system containing both tumor-targeting groups and enzyme response sites. This system enables the targeted delivery of peptide nanocarriers to tumor cells and a unique response to alkaline phosphatase (ALP) in the tumor microenvironment to activate morphological transformation and drug release. The amphiphilic peptide AYR self-aggregated into a spherical nanoparticle structure after encapsulating the lipid-soluble model drug doxorubicin (DOX) and rapidly converted to nanofibers via the induction of ALP. This morphological transformation toward a high aspect ratio allowed rapid, as well as effective drug release to tumor location while enhancing specific toxicity to tumor cells. Interestingly, this "transformer"-like drug delivery strategy can enhance local drug accumulation and effectively inhibit drug efflux. In vitro along with in vivo experiments further proved that the permeability and retention of antitumor drugs in tumor cells and tissues were significantly enhanced to reduce toxic side effects, and the therapeutic effect was remarkably improved compared with that of nondeformable drug-loaded peptide nanocarriers. The developed AYR nanoparticles with the ability to undergo morphological transformation in situ can improve local drug aggregation and retention time at the tumor site. Our findings provide a new and simple method for nanocarrier morphology transformation in novel cancer treatments.
Collapse
Affiliation(s)
- Zhongying Gong
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Xiaoying Liu
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Juanjuan Cao
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Zexin Hong
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Jingye Wang
- Department of Pathology, Weifang Maternal and Child Health Hospital, Weifang 261000, China
| | - Xirui Sun
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Xiaomeng Yuan
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Hongjie Ji
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
28
|
Destabilization of the Alzheimer's amyloid-β peptide by a proline-rich β-sheet breaker peptide: a molecular dynamics simulation study. J Mol Model 2021; 27:356. [PMID: 34796404 DOI: 10.1007/s00894-021-04968-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/25/2021] [Indexed: 11/27/2022]
Abstract
The amyloid-β peptide exists in the form of fibrils in the plaques found in the brains of patients with Alzheimer's disease. One of the therapeutic strategies is the design of molecules which can destabilize these fibrils. We present a designed peptide KLVFFP5 with two segments: the self-recognition sequence KLVFF and a β-sheet breaker proline pentamer. Molecular dynamics simulations and docking results showed that this peptide could bind to the protofibrils and destabilize them by establishing hydrophobic contacts and hydrogen bonds with a higher affinity than the KLVFF peptide. In the presence of the KLVFFP5 peptide, the β-sheet content of the protofibrils was reduced significantly; the hydrogen bonding network and the salt bridges were disrupted to a greater extent than the KLVFF peptide. Our results indicate that the KLVFFP5 peptide is an effective β-sheet disruptor which can be considered in the therapy of Alzheimer's disease.
Collapse
|
29
|
Wan W, Zeng L, Jin W, Chen X, Shen D, Huang Y, Wang M, Bai Y, Lyu H, Dong X, Gao Z, Wang L, Liu X, Liu Y. A Solvatochromic Fluorescent Probe Reveals Polarity Heterogeneity upon Protein Aggregation in Cells. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Lianggang Zeng
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Xinxin Chen
- National Laboratory of Biomacromolecules CAS Center for Excellence in Biomacromolecules Institute of Biophysics Chinese Academy of Sciences 15 Datun Road, Chaoyang District Beijing 100101 China
| | - Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Yanan Huang
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Mengdie Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Yulong Bai
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Haochen Lyu
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University 467 Zhongshan Road 116023 China Dalian
| | - Zhenming Gao
- The Second Hospital of Dalian Medical University 467 Zhongshan Road 116023 China Dalian
| | - Lei Wang
- National Laboratory of Biomacromolecules CAS Center for Excellence in Biomacromolecules Institute of Biophysics Chinese Academy of Sciences 15 Datun Road, Chaoyang District Beijing 100101 China
| | - Xiaojing Liu
- Institute of Molecular Sciences and Engineering Shan Dong University 72 Binhai Road Qingdao 266237 China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| |
Collapse
|
30
|
Hong Z, Sun X, Sun X, Cao J, Yang Z, Pan Z, Yu T, Dong J, Zhou B, Bai J. Enzyme-induced morphological transformation of drug carriers: Implications for cytotoxicity and the retention time of antitumor agents. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112389. [PMID: 34579908 DOI: 10.1016/j.msec.2021.112389] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 02/09/2023]
Abstract
Nanocarriers have been widely employed to deliver chemotherapeutic drugs for cancer treatment. However, the insufficient accumulation of nanoparticles in tumors is an important reason for the poor efficacy of nanodrugs. In this study, a novel drug delivery system with a self-assembled amphiphilic peptide was designed to respond specifically to alkaline phosphatase (ALP), a protease overexpressed in cancer cells. The amphiphilic peptide self-assembled into spherical and fibrous nanostructures, and it easily assembled into spherical drug-loaded peptide nanoparticles after loading of a hydrophobic chemotherapeutic drug. The cytotoxicity of the drug carriers was enhanced against tumor cells over time. These spherical nanoparticles transformed into nanofibers under the induction of ALP, leading to efficient release of the encapsulated drug. This drug delivery strategy relying on responsiveness to an enzyme present in the tumor microenvironment can enhance local drug accumulation at the tumor site. The results of live animal imaging showed that the residence time of the morphologically transformable drug-loaded peptide nanoparticles at the tumor site was prolonged in vivo, confirming their potential use in antitumor therapy. These findings can contribute to a better understanding of the influence of drug carrier morphology on intracellular retention.
Collapse
Affiliation(s)
- Zexin Hong
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Xirui Sun
- Department of Oncology, Weifang Medical University, Weifang 261053, China
| | - Xiumei Sun
- Department of Oncology, Weifang Medical University, Weifang 261053, China
| | - Juanjuan Cao
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Zhengqiang Yang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Zhifang Pan
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Tao Yu
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Jinhua Dong
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
31
|
Wan W, Zeng L, Jin W, Chen X, Shen D, Huang Y, Wang M, Bai Y, Lyu H, Dong X, Gao Z, Wang L, Liu X, Liu Y. A Solvatochromic Fluorescent Probe Reveals Polarity Heterogeneity upon Protein Aggregation in Cells. Angew Chem Int Ed Engl 2021; 60:25865-25871. [PMID: 34562048 DOI: 10.1002/anie.202107943] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/27/2021] [Indexed: 02/02/2023]
Abstract
We report a crystallization-induced emission fluorophore to quantitatively interrogate the polarity of aggregated proteins. This solvatochromic probe, namely "AggRetina" probe, inherently binds to aggregated proteins and exhibits both a polarity-dependent fluorescence emission wavelength shift and a viscosity-dependent fluorescence intensity increase. Regulation of its polarity sensitivity was achieved by extending the conjugation length. Different proteins bear diverse polarity upon aggregation, leading to different resistance to proteolysis. Polarity primarily decreases during protein misfolding but viscosity mainly increases upon the formation of insoluble aggregates. We quantified the polarity of aggregated protein-of-interest in live cells via HaloTag bioorthogonal labeling, revealing polarity heterogeneity within cellular aggregates. The enriched micro-environment details inside misfolded and aggregated proteins may correlate to their bio-chemical properties and pathogenicity.
Collapse
Affiliation(s)
- Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Lianggang Zeng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xinxin Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Yanan Huang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Mengdie Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Yulong Bai
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Haochen Lyu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xuepeng Dong
- The Second Hospital of, Dalian Medical University, 467 Zhongshan Road, 116023, China, Dalian
| | - Zhenming Gao
- The Second Hospital of, Dalian Medical University, 467 Zhongshan Road, 116023, China, Dalian
| | - Lei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Xiaojing Liu
- Institute of Molecular Sciences and Engineering, Shan Dong University, 72 Binhai Road, Qingdao, 266237, China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| |
Collapse
|
32
|
Xie Y, Wang Y, Jiang S, Xiang X, Wang J, Ning L. Novel strategies for the fight of Alzheimer's disease targeting amyloid-β protein. J Drug Target 2021; 30:259-268. [PMID: 34435898 DOI: 10.1080/1061186x.2021.1973482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD), which is recognised as a devastating neurodegenerative disease throughout the world and lack of effective treatments, is a growing concern in modern society with a growing population of elderly patients. A growing number of studies reveal that abnormal accumulation and deposition of Aβ is responsible for AD. Inspired by this, strategies for the treatment of AD targeting-Aβ clearance have been discussed for a long period, exploring new drugs which is capable of destroying soluble Aβ oligomers and unsolvable Aβ aggregates. In this paper, results of recent clinical trials on several anti-amyloid-β drugs are presented and several emerging anti-amyloid AD therapies based on recent studies are reviewed. Furthermore, some of the current challenges and novel strategies to prevent AD are addressed. Herein, this review focuses on current pharmacotherapy of AD targeting-Aβ and intends to design a promising therapeutic agent for AD treatment.
Collapse
Affiliation(s)
- Yang Xie
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yan Wang
- Chemistry and Chemical Engineering College, Huangshan University, Huangshan, China
| | - Shangfei Jiang
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Xiaohong Xiang
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Jianhua Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, China
| | - Linhong Ning
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| |
Collapse
|
33
|
Peng YB, Tao C, Tan CP, Zhao P. Inhibition of Aβ peptide aggregation by ruthenium(II) polypyridyl complexes through copper chelation. J Inorg Biochem 2021; 224:111591. [PMID: 34450410 DOI: 10.1016/j.jinorgbio.2021.111591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
Alzheimer's disease (AD) is known as a complex multifactorial syndrome and both metal chelators and amyloid β peptide (Aβ) inhibitors show promise against AD. Herein, four small hybrid compounds have been designed and synthesized utilizing 8-hydroxyquinoline, pyridine or imidazole as chelators and benzimidazole as the recognition moiety for AD treatment. These conjugates can capture Cu2+ from Aβ and become dimers upon Cu2+ coordination and show high efficiency for both Cu2+ elimination and Aβ assembly inhibition. Besides, these designed complexes can inhibit the production of Aβ-induced reactive oxygen species (ROS), protect mitochondria from damage, and improve the survival rate of neuron cells. Our work provides a new strategy to combine hydrophobic interaction and metal ion chelation to design amyloid inhibitors.
Collapse
Affiliation(s)
- Yan-Bo Peng
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Mega Centre, No. 280, Waihuandong Road, Guangzhou 510006, PR China
| | - Can Tao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Mega Centre, No. 280, Waihuandong Road, Guangzhou 510006, PR China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, PR China.
| | - Ping Zhao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Mega Centre, No. 280, Waihuandong Road, Guangzhou 510006, PR China.
| |
Collapse
|
34
|
Chan TG, Ruehl CL, Morse SV, Simon M, Rakers V, Watts H, Aprile FA, Choi JJ, Vilar R. Modulation of amyloid-β aggregation by metal complexes with a dual binding mode and their delivery across the blood-brain barrier using focused ultrasound. Chem Sci 2021; 12:9485-9493. [PMID: 34349923 PMCID: PMC8278877 DOI: 10.1039/d1sc02273c] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/14/2021] [Indexed: 11/21/2022] Open
Abstract
One of the key hallmarks of Alzheimer's disease is the aggregation of the amyloid-β peptide to form fibrils. Consequently, there has been great interest in studying molecules that can disrupt amyloid-β aggregation. While a handful of molecules have been shown to inhibit amyloid-β aggregation in vitro, there remains a lack of in vivo data reported due to their inability to cross the blood-brain barrier. Here, we investigate a series of new metal complexes for their ability to inhibit amyloid-β aggregation in vitro. We demonstrate that octahedral cobalt complexes with polyaromatic ligands have high inhibitory activity thanks to their dual binding mode involving π-π stacking and metal coordination to amyloid-β (confirmed via a range of spectroscopic and biophysical techniques). In addition to their high activity, these complexes are not cytotoxic to human neuroblastoma cells. Finally, we report for the first time that these metal complexes can be safely delivered across the blood-brain barrier to specific locations in the brains of mice using focused ultrasound.
Collapse
Affiliation(s)
- Tiffany G Chan
- Department of Chemistry, Imperial College London White City Campus, 82 Wood Lane London W12 0BZ UK
- Department of Bioengineering, Imperial College London London SW7 2AZ UK
- Centre of Excellence in Neurotechnology, Imperial College London London SW7 2AZ UK
| | - Carmen L Ruehl
- Department of Chemistry, Imperial College London White City Campus, 82 Wood Lane London W12 0BZ UK
| | - Sophie V Morse
- Department of Bioengineering, Imperial College London London SW7 2AZ UK
| | - Michelle Simon
- Department of Chemistry, Imperial College London White City Campus, 82 Wood Lane London W12 0BZ UK
| | - Viktoria Rakers
- Department of Chemistry, Imperial College London White City Campus, 82 Wood Lane London W12 0BZ UK
| | - Helena Watts
- Department of Bioengineering, Imperial College London London SW7 2AZ UK
- Department of Brain Sciences, Imperial College London London W12 0NN UK
| | - Francesco A Aprile
- Department of Chemistry, Imperial College London White City Campus, 82 Wood Lane London W12 0BZ UK
| | - James J Choi
- Department of Bioengineering, Imperial College London London SW7 2AZ UK
| | - Ramon Vilar
- Department of Chemistry, Imperial College London White City Campus, 82 Wood Lane London W12 0BZ UK
| |
Collapse
|
35
|
Shen D, Jin W, Bai Y, Huang Y, Lyu H, Zeng L, Wang M, Tang Y, Wan W, Dong X, Gao Z, Piao H, Liu X, Liu Y. Rational Design of Crystallization‐Induced‐Emission Probes To Detect Amorphous Protein Aggregation in Live Cells. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Yulong Bai
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Yanan Huang
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Haochen Lyu
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Lianggang Zeng
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Mengdie Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Yuqi Tang
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University 467 Zhongshan Road Dalian 116044 China
| | - Zhenming Gao
- The Second Hospital of Dalian Medical University 467 Zhongshan Road Dalian 116044 China
| | - Hai‐Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Xiaojing Liu
- Institute of Molecular Sciences and Engineering Shan Dong University Jimobinhai Road Qingdao 266237 China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry Dalian Institute of Chemical Physics Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| |
Collapse
|
36
|
Shen D, Jin W, Bai Y, Huang Y, Lyu H, Zeng L, Wang M, Tang Y, Wan W, Dong X, Gao Z, Piao HL, Liu X, Liu Y. Rational Design of Crystallization-Induced-Emission Probes To Detect Amorphous Protein Aggregation in Live Cells. Angew Chem Int Ed Engl 2021; 60:16067-16076. [PMID: 33991044 DOI: 10.1002/anie.202103674] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 12/19/2022]
Abstract
Unlike amyloid aggregates, amorphous protein aggregates with no defined structures have been challenging to target and detect in a complex cellular milieu. In this study, we rationally designed sensors of amorphous protein aggregation from aggregation-induced-emission probes (AIEgens). Utilizing dicyanoisophorone as a model AIEgen scaffold, we first sensitized the fluorescence of AIEgens to a nonpolar and viscous environment mimicking the interior of amorphous aggregated proteins. We identified a generally applicable moiety (dimethylaminophenylene) for selective binding and fluorescence enhancement. Regulation of the electron-withdrawing groups tuned the emission wavelength while retaining selective detection. Finally, we utilized the optimized probe to systematically image aggregated proteome upon proteostasis network regulation. Overall, we present a rational approach to develop amorphous protein aggregation sensors from AIEgens with controllable sensitivity, spectral coverage, and cellular performance.
Collapse
Affiliation(s)
- Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Yulong Bai
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Yanan Huang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Haochen Lyu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Lianggang Zeng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Mengdie Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Yuqi Tang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xuepeng Dong
- The Second Hospital of, Dalian Medical University, 467 Zhongshan Road, Dalian, 116044, China
| | - Zhenming Gao
- The Second Hospital of, Dalian Medical University, 467 Zhongshan Road, Dalian, 116044, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xiaojing Liu
- Institute of Molecular Sciences and Engineering, Shan Dong University, Jimobinhai Road, Qingdao, 266237, China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| |
Collapse
|
37
|
Wall BJ, Will MF, Yawson GK, Bothwell PJ, Platt DC, Apuzzo CF, Jones MA, Ferrence GM, Webb MI. Importance of Hydrogen Bonding: Structure-Activity Relationships of Ruthenium(III) Complexes with Pyridine-Based Ligands for Alzheimer's Disease Therapy. J Med Chem 2021; 64:10124-10138. [PMID: 34197109 DOI: 10.1021/acs.jmedchem.1c00360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, where one of the pathological hallmarks of AD is extracellular protein deposits, the primary component of which is the peptide amyloid-β (Aβ). Recently, the soluble form of Aβ has been recognized as the primary neurotoxic species, making it an important target for therapeutic development. Metal-based drugs are promising candidates to target Aβ, as the interactions with the peptide can be tuned by ligand design. In the current study, 11 ruthenium complexes containing pyridine-based ligands were prepared, where the functional groups at the para position on the coordinated pyridine ligand were varied to determine structure-activity relationships. Overall, the complexes with terminal primary amines had the greatest impact on modulating the aggregation of Aβ and diminishing its cytotoxicity. These results identify the importance of specific intermolecular interactions and are critical in the advancement of metal-based drugs for AD therapy.
Collapse
Affiliation(s)
- Brendan J Wall
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Mark F Will
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Gideon K Yawson
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Paige J Bothwell
- Core Microscope Facility, Department of Biological Sciences, Northern Illinois University, DeKalb, Illinois 60115, United States
| | - David C Platt
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - C Fiore Apuzzo
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Marjorie A Jones
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Gregory M Ferrence
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Michael I Webb
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| |
Collapse
|
38
|
Tong J, Wang J, Shen X, Zhang H, Wang Y, Fang Q, Chen L. One-Pot Synthesis of Schiff Bases by Defect-Induced TiO 2-x-Catalyzed Tandem Transformation from Alcohols and Nitro Compounds. Inorg Chem 2021; 60:10715-10721. [PMID: 34184890 DOI: 10.1021/acs.inorgchem.1c01406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Schiff bases that are generally formed from condensation reactions of aldehydes (or ketones) and amino groups could also be produced by a photodriven one-pot tandem reaction between alcohols and nitro compounds, in our case. Herein, TiO2-x porous cages derived from NH2-MIL-125 by a self-sacrificing template route are used to study the organic transformation and exhibit 100% conversion efficiency of nitrobenzene and 100% selectivity for Schiff bases in the system of benzyl alcohol (5 mL) and nitrobenzene (41 μL) upon light irradiation, but hydrogen by dehydrogenation of benzyl alcohol cannot be detected. Successful occurrence of the organic transformation is mainly attributed to Ti(III)-oxygen vacancy associates. Surface oxygen vacancy-related Ti(III) sites are responsible for binding with nitro groups, and low-coordinated Ti5c sites selectively adsorb hydroxyl groups of benzyl alcohol. The Ti(III) and oxygen vacancy associates capture photogenerated electrons for achievement of multielectron reduction of nitrobenzene and the subsequent Schiff base condensation reaction with the as-formed benzaldehyde.
Collapse
Affiliation(s)
- Jing Tong
- Department of Pharmaceutical Engineering, Bengbu Medical College, Bengbu, Anhui 233030, P. R. China
| | - Jinfeng Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, P. R. China
| | - Xiaoshuang Shen
- School of Physical Science & Technology, Yangzhou University, Yangzhou 225002, P. R. China
| | - Hui Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, P. R. China
| | - Yao Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, P. R. China
| | - Qiang Fang
- Department of Pharmaceutical Engineering, Bengbu Medical College, Bengbu, Anhui 233030, P. R. China
| | - Liyong Chen
- Department of Pharmaceutical Engineering, Bengbu Medical College, Bengbu, Anhui 233030, P. R. China.,State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, P. R. China
| |
Collapse
|
39
|
Aronica PGA, Reid LM, Desai N, Li J, Fox SJ, Yadahalli S, Essex JW, Verma CS. Computational Methods and Tools in Antimicrobial Peptide Research. J Chem Inf Model 2021; 61:3172-3196. [PMID: 34165973 DOI: 10.1021/acs.jcim.1c00175] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The evolution of antibiotic-resistant bacteria is an ongoing and troubling development that has increased the number of diseases and infections that risk going untreated. There is an urgent need to develop alternative strategies and treatments to address this issue. One class of molecules that is attracting significant interest is that of antimicrobial peptides (AMPs). Their design and development has been aided considerably by the applications of molecular models, and we review these here. These methods include the use of tools to explore the relationships between their structures, dynamics, and functions and the increasing application of machine learning and molecular dynamics simulations. This review compiles resources such as AMP databases, AMP-related web servers, and commonly used techniques, together aimed at aiding researchers in the area toward complementing experimental studies with computational approaches.
Collapse
Affiliation(s)
- Pietro G A Aronica
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | - Lauren M Reid
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671.,School of Chemistry, University of Southampton, Highfield Southampton, Hampshire, U.K. SO17 1BJ.,MedChemica Ltd, Alderley Park, Macclesfield, Cheshire, U.K. SK10 4TG
| | - Nirali Desai
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671.,Division of Biological and Life Sciences, Ahmedabad University, Central Campus, Ahmedabad, Gujarat, India 380009
| | - Jianguo Li
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671.,Singapore Eye Research Institute, 20 College Road Discovery Tower, Singapore 169856
| | - Stephen J Fox
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | - Shilpa Yadahalli
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | - Jonathan W Essex
- School of Chemistry, University of Southampton, Highfield Southampton, Hampshire, U.K. SO17 1BJ
| | - Chandra S Verma
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671.,Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543 Singapore.,School of Biological Sciences, Nanyang Technological University, 50 Nanyang Drive, 637551 Singapore
| |
Collapse
|
40
|
Bai Y, Wan W, Huang Y, Jin W, Lyu H, Xia Q, Dong X, Gao Z, Liu Y. Quantitative interrogation of protein co-aggregation using multi-color fluorogenic protein aggregation sensors. Chem Sci 2021; 12:8468-8476. [PMID: 34221329 PMCID: PMC8221170 DOI: 10.1039/d1sc01122g] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/19/2021] [Indexed: 12/27/2022] Open
Abstract
Co-aggregation of multiple pathogenic proteins is common in neurodegenerative diseases but deconvolution of such biochemical process is challenging. Herein, we developed a dual-color fluorogenic thermal shift assay to simultaneously report on the aggregation of two different proteins and quantitatively study their thermodynamic stability during co-aggregation. Expansion of spectral coverage was first achieved by developing multi-color fluorogenic protein aggregation sensors. Orthogonal detection was enabled by conjugating sensors of minimal fluorescence crosstalk to two different proteins via sortase-tag technology. Using this assay, we quantified shifts in melting temperatures in a heterozygous model protein system, revealing that the thermodynamic stability of wild-type proteins was significantly compromised by the mutant ones but not vice versa. We also examined how small molecule ligands selectively and differentially interfere with such interplay. Finally, we demonstrated these sensors are suited to visualize how different proteins exert influence on each other upon their co-aggregation in live cells.
Collapse
Affiliation(s)
- Yulong Bai
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Yanan Huang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Wenhan Jin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Haochen Lyu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| | - Qiuxuan Xia
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University 467 Zhongshan Road Dalian 116044 China
| | - Zhenming Gao
- The Second Hospital of Dalian Medical University 467 Zhongshan Road Dalian 116044 China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China
| |
Collapse
|
41
|
Sen N, Hause G, Binder WH. Membrane Anchored Polymers Modulate Amyloid Fibrillation. Macromol Rapid Commun 2021; 42:e2100120. [PMID: 33987913 DOI: 10.1002/marc.202100120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/26/2021] [Indexed: 12/24/2022]
Abstract
The nucleating role of cellular membrane components, such as lipid moieties on amyloid beta (Aβ1-40 ) fibrillation, has been reported in recent years. The influence of conjugates fabricated from lipid anchors (cholesterol, diacylglycerol) and hydrophilic polymers on Aβ1-40 fibrillation is reported here, aiming to understand the impact of polymers cloud point temperature (Tcp ) and its hydrophobic tails on the amyloid fibrillation. Novel lipid-polymer conjugates, consisting of poly(oligo(ethylene glycol)m acrylates) and hydrophobic groups (diacylglyceryl-, cholesteryl-, octyl-, decyl-, hexadecyl-) as anchors are synthesized using reversible addition-fragmentation chain transfer (RAFT) polymerization, allowing to tune the hydrophilic-hydrophobic profile of the conjugates by varying both, the degree of polymerization (n) and number of ethylene glycol units (m) in their side chain. The impact of these conjugates on Aβ1-40 fibrillation is investigated via in vitro kinetic studies and transmission electron microscopy (TEM). Hydrophobic lipid-anchors are significantly delaying fibrillation (both lag- and half times), observing similar fibrillar structures via TEM when compared to native Aβ1-40 . Other hydrophobic end groups are also delaying fibrillation of Aβ1-40 , irrespective of their "n" and "m," whereas more hydrophilic polymers (both with longer ethylene glycol-sidechains, m = 3 for octyl, decyl and m = 5 for cholesterol) are only marginally inhibited fibrillation.
Collapse
Affiliation(s)
- Newton Sen
- Chair of Macromolecular Chemistry, Faculty of Natural Science II, Von-Danckelmann-Platz 4, Institute of Chemistry, Martin-Luther University Halle-Wittenberg, Halle (Saale), D-06120, Germany
| | - Gerd Hause
- Biocenter, Martin-Luther University Halle-Wittenberg, Weinbergweg 22, Halle (Saale), D-06120, Germany
| | - Wolfgang H Binder
- Chair of Macromolecular Chemistry, Faculty of Natural Science II, Von-Danckelmann-Platz 4, Institute of Chemistry, Martin-Luther University Halle-Wittenberg, Halle (Saale), D-06120, Germany
| |
Collapse
|
42
|
Pasandideh S, Arasteh A. Evaluation of antioxidant and inhibitory properties of Citrus aurantium L. on the acetylcholinesterase activity and the production of amyloid nano-bio fibrils. Int J Biol Macromol 2021; 182:366-372. [PMID: 33848544 DOI: 10.1016/j.ijbiomac.2021.04.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/16/2021] [Accepted: 04/07/2021] [Indexed: 11/17/2022]
Abstract
The blossoms of Citrus aurantium are considered for the treatment of Alzheimer's disease because of their fragrant essential oils. The aim of this study was to investigate the antioxidant and inhibitory effects of Citrus aurantium extract on the acetylcholinesterase and production of amyloid nanobiofibrils from bovine serum albumin (BSA). The Citrus aurantium petals were harvested from Rasht city in northern IRAN. Chemical composition was investigated by GC-MS. The anti-Alzheimer's effects were evaluated by determining the antioxidant percentage by DPPH method and determining acetylcholinesterase activity. Congored spectroscopy was used for investigation of the inhibitory properties of the extract on the production of amyloid nanobiofibrils, and amyloid fibers was confirmed by electron microscopy. The most abundant ingredients were D-Glucuronic acid (9.53%), D-Limonene (5.54%), Linalool (2.06%), Daphnetin (3.73%), Phthalic acid (0.72%), Octadecenoic acid (3.98%), Hexadecanoic acid (2.13%), Pyrrolidinone (1.17%) and the highest antioxidant capacity was at 8 mg/ml (EC50: 2.36 mg/ml). The extract reduced the Acetylcholinesterase activity less than 47.04% (IC50: 42.8 mg/ml) and amyloid production less than 22% (EC50: 3.135). Citrus aurantium petals with inhibitory properties for the production of amyloid nanobiofibrils, can be used as a beneficial drugs for reducing side effects of Alzheimer's disease.
Collapse
Affiliation(s)
- Shayan Pasandideh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Amir Arasteh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| |
Collapse
|
43
|
Kanchi PK, Dasmahapatra AK. Enhancing the binding of the β-sheet breaker peptide LPFFD to the amyloid-β fibrils by aromatic modifications: A molecular dynamics simulation study. Comput Biol Chem 2021; 92:107471. [PMID: 33706107 DOI: 10.1016/j.compbiolchem.2021.107471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 11/25/2022]
Abstract
Alzheimer's is a fatal neurodegenerative disease for which there is no cure at present. The disease is characterized by the presence of plaques in the brains of a patient, which are composed mainly of aggregates of the amyloid-β peptide in the form of β-sheet fibrils. Here, we investigated the possibility of exploiting the superior binding ability of aromatic amino acids to a particular model of the amyloid-β fibrils. which is a difficult target for drug design. The β-sheet breaker peptide LPFFD was modified with aromatic amino acids and its binding to these fibrils was studied. We found that the orientation and the electrostatic complementarity of the modified peptide with respect to the fibrils played a crucial role in determining whether its binding was improved by the aromatic amino acids. The modified LPFFD peptides were able to bind to those fibril residues. which are important in the aggregation of amyloid-β peptides and thus can potentially inhibit the further aggregation of the amyloid-beta peptides by blocking their interactions. We found that the tryptophan modified LPFFD peptides had the best binding affinities. In most cases, the aromatic amino acids in the N-terminus of the modified peptides made more contacts with the fibrils than those in the C-terminus. We also found that increasing the aromatic content did not significantly improve the binding of the LPFFD peptide to the fibrils. Our study can serve as a basis for the design of novel peptide-based drugs for Alzheimer's disease in which aromatic interactions play an important role.
Collapse
Affiliation(s)
- Pavan Krishna Kanchi
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Ashok Kumar Dasmahapatra
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Center for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
44
|
Babu E, Bhuvaneswari J, Rajakumar K, Sathish V, Thanasekaran P. Non-conventional photoactive transition metal complexes that mediated sensing and inhibition of amyloidogenic aggregates. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213612] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
45
|
Zhou X, Wang S, Zhang C, Lin Y, Lv J, Hu S, Zhang S, Li M. Colorimetric determination of amyloid-β peptide using MOF-derived nanozyme based on porous ZnO-Co 3O 4 nanocages. Mikrochim Acta 2021; 188:56. [PMID: 33502585 DOI: 10.1007/s00604-021-04705-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/09/2021] [Indexed: 12/24/2022]
Abstract
A sensitive and rapid colorimetric biosensor has been developed for determination of amyloid-β peptide (Aβ) and study of amyloidogenesis based on the high peroxidase-like activity of porous bimetallic ZnO-Co3O4 nanocages (NCs). Due to the high binding ability of Aβ monomer to ZnO-Co3O4 NCs, the catalytic activity of ZnO-Co3O4 NCs can be significantly suppressed by Aβ monomer. This finding forms the basis for a colorimetric assay for Aβ monomer detection. The detection limit for Aβ monomer is 3.5 nM with a linear range of 5 to 150 nM (R2 = 0.997). The system was successfully applied to the determination of Aβ monomer in rat cerebrospinal fluid. Critically, the different inhibition effects of monomeric and aggregated Aβ species on the catalytic activity of ZnO-Co3O4 NCs enabled the sensor to be used for tracking the dynamic progress of Aβ aggregation and screening Aβ inhibitors. Compared with the commonly used thioflavin T fluorescence assay, this method provided higher sensitivity to the formation of Aβ oligomer at the very early assembly stage. Our assay shows potential application in early diagnosis and therapy of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Xi Zhou
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuangling Wang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Cong Zhang
- Department of Chemistry, School of Sciences, Hebei University of Science and Technology, Shijiazhuang, 050018, China.
| | - Yulong Lin
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jie Lv
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuyang Hu
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shanshan Zhang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Meng Li
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
46
|
Synthesis, spectral characterization, DNA/BSA binding, antimicrobial and in vitro cytotoxicity of cobalt(III) complexes containing 7-hydroxy-4-oxo-4H-chromene Schiff bases. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2020.120060] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
47
|
Chan KH, Lim J, Jee JE, Aw JH, Lee SS. Peptide-Peptide Co-Assembly: A Design Strategy for Functional Detection of C-peptide, A Biomarker of Diabetic Neuropathy. Int J Mol Sci 2020; 21:ijms21249671. [PMID: 33352955 PMCID: PMC7766332 DOI: 10.3390/ijms21249671] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022] Open
Abstract
Diabetes-related neuropathy is a debilitating condition that may be averted if it can be detected early. One possible way this can be achieved at low cost is to utilise peptides to detect C-peptide, a biomarker of diabetic neuropathy. This depends on peptide-peptide co-assembly, which is currently in a nascent stage of intense study. Instead, we propose a bead-based triple-overlay combinatorial strategy that can preserve inter-residue information during the screening process for a suitable complementary peptide to co-assemble with C-peptide. The screening process commenced with a pentapeptide general library, which revealed histidine to be an essential residue. Further screening with seven tetrapeptide focused libraries led to a table of self-consistent peptide sequences that included tryptophan and lysine at high frequencies. Three complementary nonapeptides (9mer com-peptides), wpkkhfwgq (Trp-D), kwkkhfwgq (Lys-D), and KWKKHFWGQ (Lys-L) (as a negative control) were picked from this table for co-assembly studies with C-peptide. Attenuated total reflectance Fourier transform infrared (ATR-FTIR) and circular dichroism (CD) spectroscopies were utilized to study inter-peptide interactions and changes in secondary structures respectively. ATR-FTIR studies showed that there is indeed inter-peptide interaction between C-peptide and the tryptophan residues of the 9mer com-peptides. CD studies of unaggregated and colloidal C-peptide with the 9mer com-peptides suggest that the extent of co-assembly of C-peptide with Trp-D is greatest, followed by Lys-D and Lys-L. These results are promising and indicate that the presented strategy is viable for designing and evaluating longer complementary peptides, as well as complementary peptides for co-assembly with other polypeptides of interest and importance. We discuss the possibility of designing complementary peptides to inhibit toxic amyloidosis with this approach.
Collapse
Affiliation(s)
- Kiat Hwa Chan
- Division of Science, Yale-NUS College, 16 College Avenue West, Singapore 138527, Singapore;
- Correspondence: (K.H.C.); (S.S.L.)
| | - Jaehong Lim
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore; (J.L.); (J.E.J.)
| | - Joo Eun Jee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore; (J.L.); (J.E.J.)
| | - Jia Hui Aw
- Division of Science, Yale-NUS College, 16 College Avenue West, Singapore 138527, Singapore;
| | - Su Seong Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore; (J.L.); (J.E.J.)
- Correspondence: (K.H.C.); (S.S.L.)
| |
Collapse
|
48
|
Roberts KF, Brue CR, Preston A, Baxter D, Herzog E, Varelas E, Meade TJ. Cobalt(III) Schiff base complexes stabilize non-fibrillar amyloid-β aggregates with reduced toxicity. J Inorg Biochem 2020; 213:111265. [PMID: 33059154 DOI: 10.1016/j.jinorgbio.2020.111265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
The aggregation of amyloid-β (Aβ) is believed to be foundational to the pathogenesis of Alzheimer's disease (AD). In vitro aggregation kinetics have been shown to correlate with rates of disease progression in both AD patients and animal models, thus proving to be a useful metric for testing Aβ-targeted therapeutics. Here we present evidence of cobalt(III) Schiff base complex ([Co(acetylacetonate)(NH3)2]Cl; Co(III)-sb) modulation of Aβ aggregation kinetics by a variety of complementary techniques. These include Thioflavin T (ThT) fluorescence, circular dichroism (CD) spectroscopy, transmission electron microscopy (TEM), and atomic force microscopy (AFM). Our data was fitted to kinetic rate laws using a mathematical model developed by Knowles et al. in order to extract mechanistic information about the effect of Co(III)-sb on aggregation kinetics. Our analysis revealed that Co(III)-sb alters Aβ aggregation by decreasing the polymerization rate and increasing the nucleation rate, suggesting that Co(III)-sb causes Aβ to rapidly stabilize oligomeric species with reduced elongation into mature fibrils. This result was corroborated by TEM and AFM of Aβ aggregates in vitro. We also demonstrate that Aβ aggregate mixtures produced in the presence of Co(III)-sb exhibit decreased cytotoxicity compared to untreated samples.
Collapse
Affiliation(s)
- Kaleigh F Roberts
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL 60208, United States
| | - Christopher R Brue
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL 60208, United States
| | - Anna Preston
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL 60208, United States
| | - Damonick Baxter
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL 60208, United States
| | - Emma Herzog
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL 60208, United States
| | - Eleni Varelas
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL 60208, United States
| | - Thomas J Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, Evanston, IL 60208, United States.
| |
Collapse
|
49
|
Metal complexes that bind to the amyloid-β peptide of relevance to Alzheimer’s disease. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213255
expr 886172045 + 931245952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
50
|
Gomes LM, Bataglioli JC, Storr T. Metal complexes that bind to the amyloid-β peptide of relevance to Alzheimer’s disease. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|