1
|
Bujalance-Fernández J, Carro E, Jurado-Sánchez B, Escarpa A. Biocatalytic ZIF-8 surface-functionalized micromotors navigating in the cerebrospinal fluid: toward Alzheimer management. NANOSCALE 2024. [PMID: 39469769 DOI: 10.1039/d4nr02044h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Alzheimer's disease (AD) is the major cause of irreversible dementia in the elderly population worldwide and one of the major causes of the decrease in the quality of life. Efficient diagnosis and monitoring would allow a fast treatment to delay the appearance of symptoms. Herein, zeolitic imidazole framework (ZIF-8)@Au@catalase micromotors are described for motion-based sensing of copper as a marker of AD. The synthesis design was based on enzyme covalent immobilization instead of encapsulation to maximize the contact with the sample at the microscale for the potential use of extremely low AD-diagnosed sample volumes. The micromotors are prepared by asymmetric modification of ZIF-8 with a gold layer for functionalization of catalase as a compatible biocatalyst. The micromotors can propel at speeds of up to 287 ± 41 μm s-1 in cerebrospinal fluid (CSF) samples of healthy volunteers. Yet, in the presence of copper, catalase poisoning results in a decrease in the speed that can be monitored for motion-based sensing detection, as illustrated in the analysis of CSF samples from AD patients from mild to severe stages (Braak III to Braak VI). The copper-mediated modulation of catalase activity proposed here as an indicator of progression states in AD disease possesses distinct advantages such as ultrafast analysis (less than 1 min) and requiring only 1 μL of sample, holding considerable promise as a supporting prescreening tool for fast diagnosis of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- J Bujalance-Fernández
- Department of Analytical Chemistry, Physical Chemistry, and Chemical Engineering, Universidad de Alcala, Alcala de Henares, E-28802 Madrid, Spain.
| | - E Carro
- Chronic Disease Programme, UFIEC, Carlos III Health Institute, Majadahonda, Madrid, 28220, Spain
- CIBERNED, Madrid, Spain
| | - B Jurado-Sánchez
- Department of Analytical Chemistry, Physical Chemistry, and Chemical Engineering, Universidad de Alcala, Alcala de Henares, E-28802 Madrid, Spain.
- Chemical Research Institute "Andres M. del Rio", Universidad de Alcala, E-28802, Madrid, Spain
| | - A Escarpa
- Department of Analytical Chemistry, Physical Chemistry, and Chemical Engineering, Universidad de Alcala, Alcala de Henares, E-28802 Madrid, Spain.
- Chemical Research Institute "Andres M. del Rio", Universidad de Alcala, E-28802, Madrid, Spain
| |
Collapse
|
2
|
Gómez-Castro CZ, Quintanar L, Vela A. An N-terminal acidic β-sheet domain is responsible for the metal-accumulation properties of amyloid-β protofibrils: a molecular dynamics study. J Biol Inorg Chem 2024; 29:407-425. [PMID: 38811408 PMCID: PMC11186886 DOI: 10.1007/s00775-024-02061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/10/2024] [Indexed: 05/31/2024]
Abstract
The influence of metal ions on the structure of amyloid- β (Aβ) protofibril models was studied through molecular dynamics to explore the molecular mechanisms underlying metal-induced Aβ aggregation relevant in Alzheimer's disease (AD). The models included 36-, 48-, and 188-mers of the Aβ42 sequence and two disease-modifying variants. Primary structural effects were observed at the N-terminal domain, as it became susceptible to the presence of cations. Specially when β-sheets predominate, this motif orients N-terminal acidic residues toward one single face of the β-sheet, resulting in the formation of an acidic region that attracts cations from the media and promotes the folding of the N-terminal region, with implications in amyloid aggregation. The molecular phenotype of the protofibril models based on Aβ variants shows that the AD-causative D7N mutation promotes the formation of N-terminal β-sheets and accumulates more Zn2+, in contrast to the non-amyloidogenic rodent sequence that hinders the β-sheets and is more selective for Na+ over Zn2+ cations. It is proposed that forming an acidic β-sheet domain and accumulating cations is a plausible molecular mechanism connecting the elevated affinity and concentration of metals in Aβ fibrils to their high content of β-sheet structure at the N-terminal sequence.
Collapse
Affiliation(s)
- Carlos Z Gómez-Castro
- Conahcyt-Universidad Autónoma del Estado de Hidalgo, Km 4.5 Carr. Pachuca-Tulancingo, Mineral de La Reforma, 42184, Hidalgo, Mexico.
| | - Liliana Quintanar
- Department of Chemistry, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX, San Pedro Zacatenco, 07360, Gustavo A. Madero, Mexico.
| | - Alberto Vela
- Department of Chemistry, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX, San Pedro Zacatenco, 07360, Gustavo A. Madero, Mexico.
| |
Collapse
|
3
|
Żygowska J, Orlikowska M, Zhukov I, Bal W, Szymańska A. Copper interaction with cystatin C: effects on protein structure and oligomerization. FEBS J 2024; 291:1974-1991. [PMID: 38349797 DOI: 10.1111/febs.17092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 01/09/2024] [Accepted: 02/01/2024] [Indexed: 02/15/2024]
Abstract
Human cystatin C (hCC), a small secretory protein, has gained attention beyond its classical role as a cysteine protease inhibitor owing to its potential involvement in neurodegenerative disorders. This study investigates the interaction between copper(II) ions [Cu(II)] and hCC, specifically targeting histidine residues known to participate in metal binding. Through various analytical techniques, including mutagenesis, circular dichroism, fluorescence assays, gel filtration chromatography, and electron microscopy, we evaluated the impact of Cu(II) ions on the structure and oligomerization of hCC. The results show that Cu(II) does not influence the secondary and tertiary structure of the studied hCC variants but affects their stability. To explore the Cu(II)-binding site, nuclear magnetic resonance (NMR) and X-ray studies were conducted. NMR experiments revealed notable changes in signal intensities and linewidths within the region 86His-Asp-Gln-Pro-His90, suggesting its involvement in Cu(II) coordination. Both histidine residues from this fragment were found to serve as a primary anchor of Cu(II) in solution, depending on the structural context and the presence of other Cu(II)-binding agents. The presence of Cu(II) led to significant destabilization and altered thermal stability of the wild-type and H90A variant, confirming differentiation between His residues in Cu(II) binding. In conclusion, this study provides valuable insights into the interaction between Cu(II) and hCC, elucidating the impact of copper ions on protein stability and identifying potential Cu(II)-binding residues. Understanding these interactions enhances our knowledge of the role of copper in neurodegenerative disorders and may facilitate the development of therapeutic strategies targeting copper-mediated processes in protein aggregation and associated pathologies.
Collapse
Affiliation(s)
- Justyna Żygowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Marta Orlikowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Igor Zhukov
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Poland
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Poland
| | - Aneta Szymańska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| |
Collapse
|
4
|
Stehle J, Hülsmann M, Godt A, Drescher M, Azarkh M. Evaluation of Copper(II) Transfer between Amyloid-beta Peptides by Relaxation-Induced Dipolar Modulation Enhancement (RIDME). Chemphyschem 2024; 25:e202300928. [PMID: 38285014 DOI: 10.1002/cphc.202300928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 01/30/2024]
Abstract
In the brains of Alzheimer's disease patients, fibrillar aggregates containing amyloid-beta (Aβ) peptides are found, along with elevated concentrations of Cu(II) ions. The aggregation pathways of Aβ peptides can be modulated by Cu(II) ions and is determined by the formation and nature of the Cu(II)-Aβ complex. If spin-labeled, the Cu(II)-Aβ complex contains two dipolar coupled paramagnetic centers, the spin label and the Cu(II) ion. Measurement of the dipolar coupling between these paramagnetic centers by relaxation-induced dipolar modulation enhancement (RIDME) allows to monitor the complex formation and thus opens a way to follow the Cu(II) transfer between peptides if a mixture of wild-type and spin-labeled ones is used. We evaluate this approach for a specific Cu(II)-Aβ complex, the aggregation-inert Component II. The kinetics of the Cu(II) transfer can be resolved by performing RIDME in a time-dependent manner. A temporal resolution of seconds has been achieved, with the potential to reach milliseconds, using a rapid-freeze quench device to stop the Cu(II) transfer in solution after defined incubation times.
Collapse
Affiliation(s)
- Juliane Stehle
- Department of Chemistry and Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Miriam Hülsmann
- Faculty of Chemistry and Center for Molecular Materials (CM2), Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Adelheid Godt
- Faculty of Chemistry and Center for Molecular Materials (CM2), Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Malte Drescher
- Department of Chemistry and Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Mykhailo Azarkh
- Department of Chemistry and Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| |
Collapse
|
5
|
Tian Y, Shang Q, Liang R, Viles JH. Copper(II) Can Kinetically Trap Arctic and Italian Amyloid-β 40 as Toxic Oligomers, Mimicking Cu(II) Binding to Wild-Type Amyloid-β 42: Implications for Familial Alzheimer's Disease. JACS AU 2024; 4:578-591. [PMID: 38425915 PMCID: PMC10900208 DOI: 10.1021/jacsau.3c00687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
The self-association of amyloid-β (Aβ) peptide into neurotoxic oligomers is believed to be central to Alzheimer's disease (AD). Copper is known to impact Aβ assembly, while disrupted copper homeostasis impacts phenotype in Alzheimer's models. Here we show the presence of substoichiometric Cu(II) has very different impacts on the assembly of Aβ40 and Aβ42 isoforms. Globally fitting microscopic rate constants for fibril assembly indicates copper will accelerate fibril formation of Aβ40 by increasing primary nucleation, while seeding experiments confirm that elongation and secondary nucleation rates are unaffected by Cu(II). In marked contrast, Cu(II) traps Aβ42 as prefibrillar oligomers and curvilinear protofibrils. Remarkably, the Cu(II) addition to preformed Aβ42 fibrils causes the disassembly of fibrils back to protofibrils and oligomers. The very different behaviors of the two Aβ isoforms are centered around differences in their fibril structures, as highlighted by studies of C-terminally amidated Aβ42. Arctic and Italian familiar mutations also support a key role for fibril structure in the interplay of Cu(II) with Aβ40/42 isoforms. The Cu(II) dependent switch in behavior between nonpathogenic Aβ40 wild-type and Aβ40 Arctic or Italian mutants suggests heightened neurotoxicity may be linked to the impact of physiological Cu(II), which traps these familial mutants as oligomers and curvilinear protofibrils, which cause membrane permeability and Ca(II) cellular influx.
Collapse
Affiliation(s)
- Yao Tian
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - Qi Shang
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - Ruina Liang
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - John H. Viles
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| |
Collapse
|
6
|
Zeng Y, Qian S, Cao Y, Xiao W. Unravelling the complex interplay of cuproptosis, lncRNAs, and immune infiltration in Alzheimer's disease: a step towards novel therapeutic targets. Ann Hum Biol 2024; 51:2342531. [PMID: 38771661 DOI: 10.1080/03014460.2024.2342531] [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: 01/11/2024] [Accepted: 03/27/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Cuproptosis, a type of cell death involving copper ion accumulation and oxidative stress, has been implicated in the development of Alzheimer's disease (AD). AIM This study aimed to explore the potential mechanisms and roles of cuproptosis-related genes (CRGs), long non-coding RNAs (lncRNAs), and immune cells in the development of cuproptosis in AD. SUBJECTS AND METHODS Gene expression profiles of AD were acquired from the Gene Expression Omnibus (GEO) database, and differential analysis was conducted to identify CRGs. Random Forest (RF) modelling was employed to select the most crucial CRGs, which were subsequently validated in the test set. A nomogram model was created to predict AD risk and categorise AD subtypes based on the identified CRGs. A lncRNA-related ceRNA network was built, and immune cell infiltration analysis was conducted. RESULTS Twelve differentially expressed CRGs were identified in the AD dataset. The RF model pinpointed the five most critical CRGs, which were validated in the test set with an AUC of 0.90. A lncRNA-related ceRNA network was developed, and immune cell infiltration analysis revealed high levels of M1 macrophages and mast cells, along with low levels of memory B cells in AD samples. Correlation analysis unveiled associations between CRGs, lncRNAs, and differentially infiltrating immune cells. CONCLUSION This research offers insights into the potential mechanisms and roles of CRGs, lncRNAs, and immune cells in the development of cuproptosis in AD. The identified CRGs and lncRNAs may serve as potential therapeutic targets for AD, and the nomogram model may assist in early AD diagnosis and subtyping.
Collapse
Affiliation(s)
- Yi Zeng
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Siqi Qian
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yuan Cao
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wenbiao Xiao
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Bernard PJ, Bellili D, Ismaili L. Calcium channel blockers' contribution to overcoming Current drug discovery challenges in Alzheimer's disease. Expert Opin Drug Discov 2024; 19:21-32. [PMID: 37800853 DOI: 10.1080/17460441.2023.2266994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive, irreversible, and multifactorial brain disorder that gradually and insidiously destroys individual's memory, thinking, and other cognitive abilities. AREAS COVERED In this perspective, the authors examine the complex and multifactorial nature of Alzheimer's disease and believe that the best approach to develop new drugs is the MTDL strategy, which obviously faces several challenges. These challenges include identifying the key combination of targets and their suitability for coordinated actions, as well as developing an acceptable pharmacokinetic and toxicological profile to deliver a drug candidate. EXPERT OPINION Since calcium plays a crucial role in the pathology of AD, a polypharmacological approach with calcium channel blockers reinforced by activities targeting other factors involved in AD is a serious option in our opinion. This is exemplified by a phase III clinical trial using a drug combination approach with Losartan, Amlodipine (a calcium channel blocker), and Atorvastatin, as well as several MTDL-based calcium channel blockade approaches with a promising in vitro and in vivo profile.
Collapse
Affiliation(s)
- Paul J Bernard
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| | - Djamila Bellili
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| | - Lhassane Ismaili
- Université de Franche-Comté, LINC, UFR Santé, Pôle de Chimie Médicinale, Besançon, France
| |
Collapse
|
8
|
Syed RA, Hayat M, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Kaleem I, Bashir S. Aging-Related Protein Alterations in the Brain. J Alzheimers Dis 2024; 99:S5-S22. [PMID: 38339930 DOI: 10.3233/jad-230801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Aging is an intrinsic aspect of an organism's life cycle and is characterized by progressive physiological decline and increased susceptibility to mortality. Many age-associated disorders, including neurological disorders, are most commonly linked with the aging process, such as Alzheimer's disease (AD). This review aims to provide a comprehensive overview of the effects of aging and AD on the molecular pathways and levels of different proteins in the brain, including metalloproteins, neurotrophic factors, amyloid proteins, and tau proteins. AD is caused by the aggregation of amyloid proteins in the brain. Factors such as metal ions, protein ligands, and the oligomerization state of amyloid precursor protein significantly influence the proteolytic processing of amyloid-β protein precursor (AβPP). Tau, a disordered cytosolic protein, serves as the principal microtubule-associated protein in mature neurons. AD patients exhibit decreased levels of nerve growth factor within their nervous systems and cerebrospinal fluid. Furthermore, a significant increase in brain-derived neurotrophic factor resulting from the neuroprotective effect of glial cell line-derived neurotrophic factor suggests that the synergistic action of these proteins plays a role in inhibiting neuronal degeneration and atrophy. The mechanism through which Aβ and AβPP govern Cu2+ transport and their influence on Cu2+ and other metal ion pools requires elucidation in future studies. A comprehensive understanding of the influence of aging and AD on molecular pathways and varying protein levels may hold the potential for the development of novel diagnostic and therapeutic methods for the treatment of AD.
Collapse
Affiliation(s)
- Rafay Ali Syed
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Mahnoor Hayat
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Imdad Kaleem
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
9
|
Hnath B, Chen J, Reynolds J, Choi E, Wang J, Zhang D, Sha CM, Dokholyan NV. Big versus small: The impact of aggregate size in disease. Protein Sci 2023; 32:e4686. [PMID: 37243896 PMCID: PMC10273386 DOI: 10.1002/pro.4686] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Protein aggregation results in an array of different size soluble oligomers and larger insoluble fibrils. Insoluble fibrils were originally thought to cause neuronal cell deaths in neurodegenerative diseases due to their prevalence in tissue samples and disease models. Despite recent studies demonstrating the toxicity associated with soluble oligomers, many therapeutic strategies still focus on fibrils or consider all types of aggregates as one group. Oligomers and fibrils require different modeling and therapeutic strategies, targeting the toxic species is crucial for successful study and therapeutic development. Here, we review the role of different-size aggregates in disease, and how factors contributing to aggregation (mutations, metals, post-translational modifications, and lipid interactions) may promote oligomers opposed to fibrils. We review two different computational modeling strategies (molecular dynamics and kinetic modeling) and how they are used to model both oligomers and fibrils. Finally, we outline the current therapeutic strategies targeting aggregating proteins and their strengths and weaknesses for targeting oligomers versus fibrils. Altogether, we aim to highlight the importance of distinguishing the difference between oligomers and fibrils and determining which species is toxic when modeling and creating therapeutics for protein aggregation in disease.
Collapse
Affiliation(s)
- Brianna Hnath
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jiaxing Chen
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Joshua Reynolds
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Esther Choi
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jian Wang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Dongyan Zhang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Congzhou M. Sha
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
| | - Nikolay V. Dokholyan
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of Biochemistry & Molecular BiologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of ChemistryPenn State UniversityUniversity ParkPennsylvaniaUSA
| |
Collapse
|
10
|
Cordeiro LM, Soares MV, da Silva AF, Dos Santos LV, de Souza LI, da Silveira TL, Baptista FBO, de Oliveira GV, Pappis C, Dressler VL, Arantes LP, Zheng F, Soares FAA. Toxicity of Copper and Zinc alone and in combination in Caenorhabditis elegans model of Huntington's disease and protective effects of rutin. Neurotoxicology 2023:S0161-813X(23)00085-2. [PMID: 37302585 DOI: 10.1016/j.neuro.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/13/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
Copper (Cu) and Zinc (Zn) are required in small concentrations for metabolic functions, but are also toxic. There is a great concern about soil pollution by heavy metals, which may exposure the population to these toxicants, either by inhalation of dust or exposure to toxicants through ingestion of food derived from contaminated soils. In addition, the toxicity of metals in combination is questionable, as soil quality guidelines only assess them separately. It is well known that metal accumulation is often found in the pathologically affected regions of many neurodegenerative diseases, including Huntington's disease (HD). HD is caused by an autosomal dominantly inherited CAG trinucleotide repeat expansion in the huntingtin (HTT) gene. This results in the formation of a mutant huntingtin (mHTT) protein with an abnormally long polyglutamine (polyQ) repeat. The pathology of HD results in loss of neuronal cells, motor changes, and dementia. Rutin is a flavonoid found in various food sources, and previous studies indicate it has protective effects in HD models and acts as a metal chelator. However, further studies are needed to unravel its effects on metal dyshomeostasis and to discern the underlying mechanisms. In the present study, we investigated the toxic effects of long-term exposure to copper, zinc, and their mixture, and the relationship with the progression of neurotoxicity and neurodegeneration in a C. elegans-based HD model. Furthermore, we investigated the effects of rutin post metal exposure. Overall, we demonstrate that chronic exposure to the metals and their mixture altered body parameters, locomotion, and developmental delay, in addition to increasing polyQ protein aggregates in muscles and neurons causing neurodegeneration. We also propose that rutin has protective effects acting through mechanisms involving antioxidant and chelating properties. Altogether, our data provides new indications about the higher toxicity of metals in combination, the chelating potential of rutin in the C. elegans model of HD and possible strategies for future treatments of neurodegenerative diseases caused by the aggregation of proteins related to metals.
Collapse
Affiliation(s)
- Larissa Marafiga Cordeiro
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Marcell Valandro Soares
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Aline Franzen da Silva
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Luiza Venturini Dos Santos
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Larissa Ilha de Souza
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Tássia Limana da Silveira
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Fabiane Bicca Obetine Baptista
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Gabriela Vitória de Oliveira
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Cristiane Pappis
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Chemistry, Santa Maria, RS, Brazil
| | - Valderi Luiz Dressler
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Chemistry, Santa Maria, RS, Brazil
| | - Leticia Priscilla Arantes
- State University of Minas Gerais, Department of Biomedical Sciences and Health, Zip code 37900-106, Passos, MG, Brazil
| | - Fuli Zheng
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Felix Alexandre Antunes Soares
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
11
|
Silwane B, Wilson M, Kataky R. An Electrochemistry and Computational Study at an Electrified Liquid-Liquid Interface for Studying Beta-Amyloid Aggregation. MEMBRANES 2023; 13:584. [PMID: 37367788 DOI: 10.3390/membranes13060584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023]
Abstract
Amphiphilic peptides, such as Aß amyloids, can adsorb at an interface between two immiscible electrolyte solutions (ITIES). Based on previous work (vide infra), a hydrophilic/hydrophobic interface is used as a simple biomimetic system for studying drug interactions. The ITIES provides a 2D interface to study ion-transfer processes associated with aggregation, as a function of Galvani potential difference. Here, the aggregation/complexation behaviour of Aβ(1-42) is studied in the presence of Cu (II) ions, together with the effect of a multifunctional peptidomimetic inhibitor (P6). Cyclic and differential pulse voltammetry proved to be particularly sensitive to the detection of the complexation and aggregation of Aβ(1-42), enabling estimations of changes in lipophilicity upon binding to Cu (II) and P6. At a 1:1 ratio of Cu (II):Aβ(1-42), fresh samples showed a single DPV (Differential Pulse Voltammetry) peak half wave transfer potential (E1/2) at 0.40 V. Upon increasing the ratio of Cu (II) two-fold, fluctuations were observed in the DPVs, indicating aggregation. The approximate stoichiometry and binding properties of Aβ(1-42) during complexation with Cu (II) were determined by performing a differential pulse voltammetry (DPV) standard addition method, which showed two binding regimes. A pKa of 8.1 was estimated, with a Cu:Aβ1-42 ratio~1:1.7. Studies using molecular dynamics simulations of peptides at the ITIES show that Aβ(1-42) strands interact through the formation of β-sheet stabilised structures. In the absence of copper, binding/unbinding is dynamic, and interactions are relatively weak, leading to the observation of parallel and anti-parallel arrangements of β-sheet stabilised aggregates. In the presence of copper ions, strong binding occurs between a copper ion and histidine residues on two peptides. This provides a convenient geometry for inducing favourable interactions between folded β-sheet structures. Circular Dichroism spectroscopy (CD spectroscopy) was used to support the aggregation behaviour of the Aβ(1-42) peptides following the addition of Cu (II) and P6 to the aqueous phase.
Collapse
Affiliation(s)
- Bongiwe Silwane
- Department of Chemistry, Durham University, Durham DH1 3LE, UK
| | - Mark Wilson
- Department of Chemistry, Durham University, Durham DH1 3LE, UK
| | - Ritu Kataky
- Department of Chemistry, Durham University, Durham DH1 3LE, UK
| |
Collapse
|
12
|
Pal I, Dey SG. The Role of Heme and Copper in Alzheimer's Disease and Type 2 Diabetes Mellitus. JACS AU 2023; 3:657-681. [PMID: 37006768 PMCID: PMC10052274 DOI: 10.1021/jacsau.2c00572] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 06/19/2023]
Abstract
Beyond the well-explored proposition of protein aggregation or amyloidosis as the central event in amyloidogenic diseases like Alzheimer's Disease (AD), and Type 2 Diabetes Mellitus (T2Dm); there are alternative hypotheses, now becoming increasingly evident, which suggest that the small biomolecules like redox noninnocent metals (Fe, Cu, Zn, etc.) and cofactors (Heme) have a definite influence in the onset and extent of such degenerative maladies. Dyshomeostasis of these components remains as one of the common features in both AD and T2Dm etiology. Recent advances in this course reveal that the metal/cofactor-peptide interactions and covalent binding can alarmingly enhance and modify the toxic reactivities, oxidize vital biomolecules, significantly contribute to the oxidative stress leading to cell apoptosis, and may precede the amyloid fibrils formation by altering their native folds. This perspective highlights this aspect of amyloidogenic pathology which revolves around the impact of the metals and cofactors in the pathogenic courses of AD and T2Dm including the active site environments, altered reactivities, and the probable mechanisms involving some highly reactive intermediates as well. It also discusses some in vitro metal chelation or heme sequestration strategies which might serve as a possible remedy. These findings might open up a new paradigm in our conventional understanding of amyloidogenic diseases. Moreover, the interaction of the active sites with small molecules elucidates potential biochemical reactivities that can inspire designing of drug candidates for such pathologies.
Collapse
Affiliation(s)
- Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick
Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick
Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
13
|
Zimbone S, Giuffrida ML, Sabatino G, Di Natale G, Tosto R, Consoli GML, Milardi D, Pappalardo G, Sciacca MFM. Aβ 8-20 Fragment as an Anti-Fibrillogenic and Neuroprotective Agent: Advancing toward Efficient Alzheimer's Disease Treatment. ACS Chem Neurosci 2023; 14:1126-1136. [PMID: 36857606 PMCID: PMC10020970 DOI: 10.1021/acschemneuro.2c00720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterized by a spectrum of symptoms associated with memory loss and cognitive decline with deleterious consequences in everyday life. The lack of specific drugs for the treatment and/or prevention of this pathology makes AD an ever-increasing economic and social emergency. Oligomeric species of amyloid-beta (Aβ) are recognized as the primary cause responsible for synaptic dysfunction and neuronal degeneration, playing a crucial role in the onset of the pathology. Several studies have been focusing on the use of small molecules and peptides targeting oligomeric species to prevent Aβ aggregation and toxicity. Among them, peptide fragments derived from the primary sequence of Aβ have also been used to exploit any eventual recognition abilities toward the full-length Aβ parent peptide. Here, we test the Aβ8-20 fragment which contains the self-recognizing Lys-Leu-Val-Phe-Phe sequence and lacks Arg 5 and Asp 7 and the main part of the C-terminus, key points involved in the aggregation pathway and stabilization of the fibrillary structure of Aβ. In particular, by combining chemical and biological techniques, we show that Aβ8-20 does not undergo random coil to β sheet conformational transition, does not form amyloid fibrils by itself, and is not toxic for neuronal cells. Moreover, we demonstrate that Aβ8-20 mainly interacts with the 4-11 region of Aβ1-42 and inhibits the formation of toxic oligomeric species and Aβ fibrils. Finally, our data show that Aβ8-20 protects neuron-like cells from Aβ1-42 oligomer toxicity. We propose Aβ8-20 as a promising drug candidate for the treatment of AD.
Collapse
Affiliation(s)
- Stefania Zimbone
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Maria Laura Giuffrida
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Giuseppina Sabatino
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Giuseppe Di Natale
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Rita Tosto
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Grazia M L Consoli
- Consiglio Nazionale delle Ricerche, Istituto di Chimica Biomolecolare, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Danilo Milardi
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Giuseppe Pappalardo
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Michele F M Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| |
Collapse
|
14
|
Suh JM, Kim M, Yoo J, Han J, Paulina C, Lim MH. Intercommunication between metal ions and amyloidogenic peptides or proteins in protein misfolding disorders. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
15
|
Chen LL, Fan YG, Zhao LX, Zhang Q, Wang ZY. The metal ion hypothesis of Alzheimer's disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem 2023; 131:106301. [PMID: 36455485 DOI: 10.1016/j.bioorg.2022.106301] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/13/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD), characterized by the β-amyloid protein (Aβ) deposition and tau hyperphosphorylation, is the most common dementia with uncertain etiology. The clinical trials of Aβ monoclonal antibody drugs have almost failed, giving rise to great attention on the other etiologic hypothesis regarding AD such as metal ions dysmetabolism and chronic neuroinflammation. Mounting evidence revealed that the metal ions (iron, copper, and zinc) were dysregulated in the susceptible brain regions of AD patients, which was highly associated with Aβ deposition, tau hyperphosphorylation, neuronal loss, as well as neuroinflammation. Further studies uncovered that iron, copper and zinc could not only enhance the production of Aβ but also directly bind to Aβ and tau to promote their aggregations. In addition, the accumulation of iron and copper could respectively promote ferroptosis and cuproptosis. Therefore, the metal ion chelators were recognized as promising agents for treating AD. This review comprehensively summarized the effects of metal ions on the Aβ dynamics and tau phosphorylation in the progression of AD. Furthermore, taking chronic neuroinflammation contributes to the progression of AD, we also provided a summary of the mechanisms concerning metal ions on neuroinflammation and highlighted the metal ion chelators may be potential agents to alleviate neuroinflammation under the condition of AD. Nevertheless, more investigations regarding metal ions on neuroinflammation should be taken into practice, and the effects of metal ion chelators on neuroinflammation should gain more attention. Running title: Metal chelators against neuroinflammation.
Collapse
Affiliation(s)
- Li-Lin Chen
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Qi Zhang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
16
|
The co-effect of copper and lipid vesicles on Aβ aggregation. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184082. [PMID: 36374760 DOI: 10.1016/j.bbamem.2022.184082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Both metal ions and lipid membranes have a wide distribution in amyloid plaques and play significant roles in AD pathogenesis. Although influences of different metal ions or lipid vesicles on the aggregation of Aβ peptides have been extensively studied, their combined effects are less understood. In this study, we reported a unique effect of copper ion on Aβ aggregation in the presence of lipid vesicles, different from other divalent metal ions. Cu2+ in a super stoichiometric amount leads to the rapid formation of β-sheet rich structure, containing abundant low molecular weight (LMW) oligomers. We demonstrated that oligomerization of Aβ40 induced by Cu2+ binding was an essential prerequisite for the rapid conformation transition. Overall, the finding provided a new view on the complex triple system of Aβ, copper ion and lipid vesicles, which might help understanding of Aβ pathologies.
Collapse
|
17
|
Tian Y, Viles JH. pH Dependence of Amyloid-β Fibril Assembly Kinetics: Unravelling the Microscopic Molecular Processes. Angew Chem Int Ed Engl 2022; 61:e202210675. [PMID: 36197009 PMCID: PMC9828734 DOI: 10.1002/anie.202210675] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Indexed: 11/05/2022]
Abstract
Central to Alzheimer's disease (AD) is the assembly of the amyloid-beta peptide (Aβ) into fibrils. A reduction in pH accompanying inflammation or subcellular compartments, may accelerate fibril formation as the pH approaches Aβ's isoelectric point (pI). Using global fitting of fibril formation kinetics over a range of pHs, we identify the impact net charge has on individual fibril assembly microscopic rate constants. We show that the primary nucleation has a strong pH dependence. The titration behaviour exhibits a mid-point or pKa of 7.0, close to the pKa of Aβ histidine imidazoles. Surprisingly, both the secondary nucleation and elongation rate constants are pH independent. This indicates the charge of Aβ, in particular histidine protonation, has little impact on this stage of Aβ assembly. These fundamental processes are key to understanding the forces that drive the assembly of Aβ into toxic oligomers and fibrils.
Collapse
Affiliation(s)
- Yao Tian
- Department of BiochemistrySchool of Biological and Behavioural SciencesQueen Mary University of LondonLondonUK
| | - John H. Viles
- Department of BiochemistrySchool of Biological and Behavioural SciencesQueen Mary University of LondonLondonUK
| |
Collapse
|
18
|
Tian Y, Viles JH. pH Dependence of Amyloid-β Fibril Assembly Kinetics: Unravelling the Microscopic Molecular Processes. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202210675. [PMID: 38504922 PMCID: PMC10947324 DOI: 10.1002/ange.202210675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Indexed: 11/09/2022]
Abstract
Central to Alzheimer's disease (AD) is the assembly of the amyloid-beta peptide (Aβ) into fibrils. A reduction in pH accompanying inflammation or subcellular compartments, may accelerate fibril formation as the pH approaches Aβ's isoelectric point (pI). Using global fitting of fibril formation kinetics over a range of pHs, we identify the impact net charge has on individual fibril assembly microscopic rate constants. We show that the primary nucleation has a strong pH dependence. The titration behaviour exhibits a mid-point or pK a of 7.0, close to the pK a of Aβ histidine imidazoles. Surprisingly, both the secondary nucleation and elongation rate constants are pH independent. This indicates the charge of Aβ, in particular histidine protonation, has little impact on this stage of Aβ assembly. These fundamental processes are key to understanding the forces that drive the assembly of Aβ into toxic oligomers and fibrils.
Collapse
Affiliation(s)
- Yao Tian
- Department of BiochemistrySchool of Biological and Behavioural SciencesQueen Mary University of LondonLondonUK
| | - John H. Viles
- Department of BiochemistrySchool of Biological and Behavioural SciencesQueen Mary University of LondonLondonUK
| |
Collapse
|
19
|
Picone P, Sanfilippo T, Vasto S, Baldassano S, Guggino R, Nuzzo D, Bulone D, San Biagio PL, Muscolino E, Monastero R, Dispenza C, Giacomazza D. From Small Peptides to Large Proteins against Alzheimer’sDisease. Biomolecules 2022; 12:biom12101344. [PMID: 36291553 PMCID: PMC9599460 DOI: 10.3390/biom12101344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder in the elderly. The two cardinal neuropathological hallmarks of AD are the senile plaques, which are extracellular deposits mainly constituted by beta-amyloids, and neurofibrillary tangles formed by abnormally phosphorylated Tau (p-Tau) located in the cytoplasm of neurons. Although the research has made relevant progress in the management of the disease, the treatment is still lacking. Only symptomatic medications exist for the disease, and, in the meantime, laboratories worldwide are investigating disease-modifying treatments for AD. In the present review, results centered on the use of peptides of different sizes involved in AD are presented.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto per la Ricerca e l’Innovazione Biomedica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Tiziana Sanfilippo
- Ambulatorio di Nutrizione Clinica ASP Palermo, Via G. Cusmano 24, 90141 Palermo, Italy
- Anestesia e Rianimazione, Presidio Ospedaliero “S. Cimino”, 90141 Termini Imerese, Italy
| | - Sonya Vasto
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
- Istituti Euro-Mediterranei di Scienza e Tecnologia (IEMEST), Via M. Miraglia 20, 90139 Palermo, Italy
| | - Sara Baldassano
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Rossella Guggino
- Ambulatorio di Nutrizione Clinica ASP Palermo, Via G. Cusmano 24, 90141 Palermo, Italy
- Anestesia e Rianimazione, Presidio Ospedaliero “S. Cimino”, 90141 Termini Imerese, Italy
| | - Domenico Nuzzo
- Istituto per la Ricerca e l’Innovazione Biomedica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
- Correspondence: (D.N.); (D.G.)
| | - Donatella Bulone
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Pier Luigi San Biagio
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Emanuela Muscolino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze, Bldg 6, 90128 Palermo, Italy
| | - Roberto Monastero
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Clelia Dispenza
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze, Bldg 6, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Correspondence: (D.N.); (D.G.)
| |
Collapse
|
20
|
Feng J, Tang Y, Liu J, Zhang P, Liu C, Wang L. Bio-high entropy alloys: Progress, challenges, and opportunities. Front Bioeng Biotechnol 2022; 10:977282. [PMID: 36159673 PMCID: PMC9492866 DOI: 10.3389/fbioe.2022.977282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/01/2022] [Indexed: 12/01/2022] Open
Abstract
With the continuous progress and development in biomedicine, metallic biomedical materials have attracted significant attention from researchers. Due to the low compatibility of traditional metal implant materials with the human body, it is urgent to develop new biomaterials with excellent mechanical properties and appropriate biocompatibility to solve the adverse reactions caused by long-term implantation. High entropy alloys (HEAs) are nearly equimolar alloys of five or more elements, with huge compositional design space and excellent mechanical properties. In contrast, biological high-entropy alloys (Bio-HEAs) are expected to be a new bio-alloy for biomedicine due to their excellent biocompatibility and tunable mechanical properties. This review summarizes the composition system of Bio-HEAs in recent years, introduces their biocompatibility and mechanical properties of human bone adaptation, and finally puts forward the following suggestions for the development direction of Bio-HEAs: to improve the theory and simulation studies of Bio-HEAs composition design, to quantify the influence of composition, process, post-treatment on the performance of Bio-HEAs, to focus on the loss of Bio-HEAs under actual service conditions, and it is hoped that the clinical application of the new medical alloy Bio-HEAs can be realized as soon as possible.
Collapse
Affiliation(s)
- Junyi Feng
- School of Materials Engineering, Shanghai University of Engineering Science, Shanghai, China
| | - Yujin Tang
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- *Correspondence: Yujin Tang, ; Jia Liu, ; Peilei Zhang,
| | - Jia Liu
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- *Correspondence: Yujin Tang, ; Jia Liu, ; Peilei Zhang,
| | - Peilei Zhang
- School of Materials Engineering, Shanghai University of Engineering Science, Shanghai, China
- *Correspondence: Yujin Tang, ; Jia Liu, ; Peilei Zhang,
| | - Changxi Liu
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Liqiang Wang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Aβ and Tau Interact with Metal Ions, Lipid Membranes and Peptide-Based Amyloid Inhibitors: Are These Common Features Relevant in Alzheimer’s Disease? Molecules 2022; 27:molecules27165066. [PMID: 36014310 PMCID: PMC9414153 DOI: 10.3390/molecules27165066] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 12/13/2022] Open
Abstract
In the last two decades, the amyloid hypothesis, i.e., the abnormal accumulation of toxic Aβ assemblies in the brain, has been considered the mainstream concept sustaining research in Alzheimer’s Disease (AD). However, the course of cognitive decline and AD development better correlates with tau accumulation rather than amyloid peptide deposition. Moreover, all clinical trials of amyloid-targeting drug candidates have been unsuccessful, implicitly suggesting that the amyloid hypothesis needs significant amendments. Accumulating evidence supports the existence of a series of potentially dangerous relationships between Aβ oligomeric species and tau protein in AD. However, the molecular determinants underlying pathogenic Aβ/tau cross interactions are not fully understood. Here, we discuss the common features of Aβ and tau molecules, with special emphasis on: (i) the critical role played by metal dyshomeostasis in promoting both Aβ and tau aggregation and oxidative stress, in AD; (ii) the effects of lipid membranes on Aβ and tau (co)-aggregation at the membrane interface; (iii) the potential of small peptide-based inhibitors of Aβ and tau misfolding as therapeutic tools in AD. Although the molecular mechanism underlying the direct Aβ/tau interaction remains largely unknown, the arguments discussed in this review may help reinforcing the current view of a synergistic Aβ/tau molecular crosstalk in AD and stimulate further research to mechanism elucidation and next-generation AD therapeutics.
Collapse
|
22
|
Mocanu CS, Darie-Ion L, Petre BA, Gradinaru VR, Drochioiu G. A computational study of metal ions interaction with amyloid-β 1-42 peptide structure in hyperpyrexia: Implications for Alzheimer disease. JOURNAL OF KING SAUD UNIVERSITY. SCIENCE 2022; 34:102184. [PMID: 35783243 PMCID: PMC9238029 DOI: 10.1016/j.jksus.2022.102184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 05/28/2023]
Abstract
Given the current context of the SARS-CoV-19 pandemic, among the interfering risky factors with the Aβ peptide aggregation in the brains of Alzheimer's disease (AD) patients can be hyperpyrexia and increased intracranial pressure (ICP). According to our hypothesis on the relationship between hyperpyrexia and cognitive decline in AD, two models of Aβ peptides were used in this study: the structure of AD amyloid beta-peptide and near-atomic resolution fibril structures of the Aβ peptide. Therefore, the binding templates were constructed for Aβ peptide regions able to bind 9 different metal ions. The fragment transformation method was used for the structural comparison between Aβ chains. Molecular dynamics simulation (MDS) was applied using the Nose-Poincare-Anderson equation to generate a theoretically correct NPT (isothermal-isobaric ensemble). The smallest dissimilarities were observed in the case of Cu+ binding potential followed by Co2+, both with similar variation. Structural changes have also occurred as a result of the dynamic simulation. All these changes suggest an aggravating factor in both hyperpyretic and AD conditions. Our findings suggest that elevated temperature and increased intracranial pressure rise the effect of peptide aggregation, by converting α-helix motif to β-sheet and random coil conformation, which are related to the formation of senile plaques in AD brains.
Collapse
Affiliation(s)
- Cosmin Stefan Mocanu
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| | - Laura Darie-Ion
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| | - Brindusa Alina Petre
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
- Center for Fundamental Research and Experimental Development in Translation Medicine, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot Str., 700483 Iasi, Romania
| | | | - Gabi Drochioiu
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| |
Collapse
|
23
|
Liang R, Tian Y, Viles JH. Cross-seeding of WT amyloid-β with Arctic but not Italian familial mutants accelerates fibril formation in Alzheimer's disease. J Biol Chem 2022; 298:102071. [PMID: 35643314 PMCID: PMC9243174 DOI: 10.1016/j.jbc.2022.102071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 01/21/2023] Open
Abstract
Alzheimer’s disease (AD) involves the neurotoxic self-assembly of a 40 and 42 residue peptide, Amyloid-β (Aβ). Inherited early-onset AD can be caused by single point mutations within the Aβ sequence, including Arctic (E22G) and Italian (E22K) familial mutants. These mutations are heterozygous, resulting in an equal proportion of the WT and mutant Aβ isoform expression. It is therefore important to understand how these mixtures of Aβ isoforms interact with each other and influence the kinetics and morphology of their assembly into oligomers and fibrils. Using small amounts of nucleating fibril seeds, here, we systematically monitored the kinetics of fibril formation, comparing self-seeding with cross-seeding behavior of a range of isoform mixtures of Aβ42 and Aβ40. We confirm that Aβ40(WT) does not readily cross-seed Aβ42(WT) fibril formation. In contrast, fibril formation of Aβ40(Arctic) is hugely accelerated by Aβ42(WT) fibrils, causing an eight-fold reduction in the lag-time to fibrillization. We propose that cross-seeding between the more abundant Aβ40(Arctic) and Aβ42(WT) may be important for driving early-onset AD and will propagate fibril morphology as indicated by fibril twist periodicity. This kinetic behavior is not emulated by the Italian mutant, where minimal cross-seeding is observed. In addition, we studied the cross-seeding behavior of a C-terminal-amidated Aβ42 analog to probe the coulombic charge interplay between Glu22/Asp23/Lys28 and the C-terminal carboxylate. Overall, these studies highlight the role of cross-seeding between WT and mutant Aβ40/42 isoforms, which can impact the rate and structure of fibril assembly.
Collapse
Affiliation(s)
- Ruina Liang
- School of Biological and Behavioural Sciences, Queen Mary, University of London, London, United Kingdom
| | - Yao Tian
- School of Biological and Behavioural Sciences, Queen Mary, University of London, London, United Kingdom
| | - John H Viles
- School of Biological and Behavioural Sciences, Queen Mary, University of London, London, United Kingdom.
| |
Collapse
|
24
|
Chauhan BS, Rai A, Sonkar AK, Tripathi K, Upadhyay S, Mishra L, Srikrishna S. Neuroprotective Activity of a Novel Synthetic Rhodamine-Based Hydrazone against Cu 2+-Induced Alzheimer's Disease in Drosophila. ACS Chem Neurosci 2022; 13:1566-1579. [PMID: 35476931 DOI: 10.1021/acschemneuro.2c00144] [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] [Indexed: 11/28/2022] Open
Abstract
A new rhodamine-based probe 3,5-di-tert-butylsalicylaldehyde rhodamine hydrazone (RHTB) has been synthesized and well characterized using spectroscopic techniques and single-crystal X-ray crystallography. Among several metal ions, it selectively detects Cu2+ ions as monitored by UV-Vis and emission spectral titrations. It displays "turn on" behavior owing to the opening of a spirolactum ring and the presence of 3,5-di-tert-butyl as an electron releasing group. Further, Cu2+ ions play a pivotal role in extracellular aggregation of Aβ42 peptides. So far, we know probably that there are no promising drugs available in this regard. Hence, countering the Cu2+ ions by RHTB chelation against orally administered Cu2+ ion-induced neurotoxicity in the eye tissue of Drosophila expressing human Aβ42 (amyloid-β42) has been tested. The present study involves in vivo and in silico approaches. They reveal the therapeutic potential of RHTB against Cu2+ ion-induced Aβ42 toxicity in Alzheimer's disease (AD) model of Drosophila.
Collapse
Affiliation(s)
- Brijesh Singh Chauhan
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Abhishek Rai
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Avinash Kumar Sonkar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Kamini Tripathi
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Sonal Upadhyay
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Lallan Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Saripella Srikrishna
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
25
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
26
|
Singh SK, Balendra V, Obaid AA, Esposto J, Tikhonova MA, Gautam NK, Poeggeler B. Copper-Mediated β-Amyloid Toxicity and its Chelation Therapy in Alzheimer's Disease. Metallomics 2022; 14:6554256. [PMID: 35333348 DOI: 10.1093/mtomcs/mfac018] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 03/08/2022] [Indexed: 01/10/2023]
Abstract
The link between bio-metals, Alzheimer's disease (AD), and its associated protein, amyloid-β (Aβ) is very complex and one of the most studied aspects currently. Alzheimer's disease, a progressive neurodegenerative disease, is proposed to occurs due to the misfolding and aggregation of Aβ. Dyshomeostasis of metal ions and their interaction with Aβ has largely been implicated in AD. Copper plays a crucial role in amyloid-β toxicity and AD development potentially occurs through direct interaction with the copper-binding motif of APP and different amino acid residues of Aβ. Previous reports suggest that high levels of copper accumulation in the AD brain result in modulation of toxic Aβ peptide levels, implicating the role of copper in the pathophysiology of AD. In this review, we explore the possible mode of copper ion interaction with Aβ which accelerates the kinetics of fibril formation and promote amyloid-β mediated cell toxicity in Alzheimer's disease and the potential use of various copper chelators in the prevention of copper-mediated Aβ toxicity.
Collapse
Affiliation(s)
- Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Lucknow-226002, India
| | - Vyshnavy Balendra
- Saint James School of Medicine, Park Ridge, Illinois, United States of America 60068
| | - Ahmad A Obaid
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Josephine Esposto
- Department of Environmental and Life Sciences, Trent University, Peterborough, Ontario, CanadaK9L 0G2
| | - Maria A Tikhonova
- Laboratory of the Experimental Models of Neurodegenerative Processes, Scientific Research Institute of Neurosciences and Medicine; Timakov st., 4, Novosibirsk, 630117, Russia
| | - Naveen Kumar Gautam
- Department of Urology and Renal Transplantation, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Burkhard Poeggeler
- Johann-Friedrich-Blumenbach-Institute for Zoology & Anthropology, Faculty of Biology and Psychology, Georg-August-University of Göttingen, Am Türmchen 3,33332 Gütersloh, Germany
| |
Collapse
|
27
|
Klose D, Vemulapalli SPB, Richman M, Rudnick S, Aisha V, Abayev M, Chemerovski M, Shviro M, Zitoun D, Majer K, Wili N, Goobes G, Griesinger C, Jeschke G, Rahimipour S. Cu 2+-Induced self-assembly and amyloid formation of a cyclic D,L-α-peptide: structure and function. Phys Chem Chem Phys 2022; 24:6699-6715. [PMID: 35234757 DOI: 10.1039/d1cp05415e] [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
In a wide spectrum of neurodegenerative diseases, self-assembly of pathogenic proteins to cytotoxic intermediates is accelerated by the presence of metal ions such as Cu2+. Only low concentrations of these early transient oligomeric intermediates are present in a mixture of species during fibril formation, and hence information on the extent of structuring of these oligomers is still largely unknown. Here, we investigate dimers as the first intermediates in the Cu2+-driven aggregation of a cyclic D,L-α-peptide architecture. The unique structural and functional properties of this model system recapitulate the self-assembling properties of amyloidogenic proteins including β-sheet conformation and cross-interaction with pathogenic amyloids. We show that a histidine-rich cyclic D,L-α-octapeptide binds Cu2+ with high affinity and selectivity to generate amyloid-like cross-β-sheet structures. By taking advantage of backbone amide methylation to arrest the self-assembly at the dimeric stage, we obtain structural information and characterize the degree of local order for the dimer. We found that, while catalytic amounts of Cu2+ promote aggregation of the peptide to fibrillar structures, higher concentrations dose-dependently reduce fibrillization and lead to formation of spherical particles, showing self-assembly to different polymorphs. For the initial self-assembly step to the dimers, we found that Cu2+ is coordinated on average by two histidines, similar to self-assembled peptides, indicating that a similar binding interface is perpetuated during Cu2+-driven oligomerization. The dimer itself is found in heterogeneous conformations that undergo dynamic exchange, leading to the formation of different polymorphs at the initial stage of the aggregation process.
Collapse
Affiliation(s)
- Daniel Klose
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland.
| | - Sahithya Phani Babu Vemulapalli
- NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany. .,Institute for Chemistry and Biology of the Marine Environment, University of Oldenburg, 26129 Oldenburg, Germany
| | - Michal Richman
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Safra Rudnick
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel. .,Bar-Ilan Institute for Technology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Vered Aisha
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Meital Abayev
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Marina Chemerovski
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Meital Shviro
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel. .,Bar-Ilan Institute for Technology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - David Zitoun
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel. .,Bar-Ilan Institute for Technology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Katharina Majer
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland.
| | - Nino Wili
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland.
| | - Gil Goobes
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Christian Griesinger
- NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany.
| | - Gunnar Jeschke
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland.
| | - Shai Rahimipour
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|
28
|
La Penna G, Morante S. Aggregates Sealed by Ions. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2340:309-341. [PMID: 35167080 DOI: 10.1007/978-1-0716-1546-1_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The chapter draws a line connecting some recent results where the role of ions is found essential in sealing more or less pre-organized assemblies of macromolecules. We draw some dots along the line that starts from the effect of the ionic atmosphere and ends with the chemical bonds formed by multivalent ions acting as bridges between macromolecules. Many of these dots involve structurally disordered peptides and disordered regions of proteins. A broad perspective of the role of multivalent ions in assisting the assembly process, shifting population in polymorphic states, and sealing protein aggregates, is suggested.
Collapse
Affiliation(s)
- Giovanni La Penna
- Institute for Chemistry of Organo-Metallic Compounds, National Research Council of Italy, Florence, Italy.
| | - Silvia Morante
- Department of Physics, University of Roma Tor Vergata, Roma, Italy
| |
Collapse
|
29
|
Sciacca MF, Naletova I, Giuffrida ML, Attanasio F. Semax, a Synthetic Regulatory Peptide, Affects Copper-Induced Abeta Aggregation and Amyloid Formation in Artificial Membrane Models. ACS Chem Neurosci 2022; 13:486-496. [PMID: 35080861 PMCID: PMC8855339 DOI: 10.1021/acschemneuro.1c00707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
![]()
Alzheimer’s
disease, the most common form of dementia, is
characterized by the aggregation of amyloid beta protein (Aβ).
The aggregation and toxicity of Aβ are strongly modulated by
metal ions and phospholipidic membranes. In particular, Cu2+ ions play a pivotal role in modulating Aβ aggregation. Although
in the last decades several natural or synthetic compounds were evaluated
as candidate drugs, to date, no treatments are available for the pathology.
Multifunctional compounds able to both inhibit fibrillogenesis, and
in particular the formation of oligomeric species, and prevent the
formation of the Aβ:Cu2+ complex are of particular
interest. Here we tested the anti-aggregating properties of a heptapeptide,
Semax, an ACTH-like peptide, which is known to form a stable complex
with Cu2+ ions and has been proven to have neuroprotective
and nootropic effects. We demonstrated through a combination of spectrofluorometric,
calorimetric, and MTT assays that Semax not only is able to prevent
the formation of Aβ:Cu2+ complexes but also has anti-aggregating
and protective properties especially in the presence of Cu2+. The results suggest that Semax inhibits fiber formation by interfering
with the fibrillogenesis of Aβ:Cu2+ complexes.
Collapse
Affiliation(s)
- Michele F.M. Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Irina Naletova
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Maria Laura Giuffrida
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Francesco Attanasio
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| |
Collapse
|
30
|
Zhou Z, Chen S, Huang Y, Gu B, Li J, Wu C, Yin P, Zhang Y, Li H. Simultaneous visualization and quantification of copper (II) ions in Alzheimer's disease by a near-infrared fluorescence probe. Biosens Bioelectron 2022; 198:113858. [PMID: 34871835 DOI: 10.1016/j.bios.2021.113858] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/10/2021] [Accepted: 11/29/2021] [Indexed: 11/25/2022]
Abstract
The abnormal accumulation of copper ions (Cu2+) is considered to be one of the pathological factors of Alzheimer's disease (AD), but the internal relationship between Cu2+ and AD progression is still not fully clear. In this work, a sensitive and selective near-infrared fluorescent copper ion probe (DDP-Cu) was designed for quantification and visualization of Cu2+ level in lysates, living cells, living zebrafish and brain tissues of drosophila and mice with AD. By using this probe, we demonstrated that the content of Cu2+ in the brains of AD mice and drosophila enhanced nearly 3.5-fold and 4-fold than that of normal mice and drosophila, respectively. More importantly, pathogenesis analysis revealed that elevated Cu2+ led to changes in factors closely associated with AD, such as the increasing of reactive oxygen species(ROS), the aggregation of amyloid-β protein (Aβ) and nerve cell cytotoxicity. These findings could promote the understanding of the roles between Cu2+ and AD.
Collapse
Affiliation(s)
- Zile Zhou
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Shengyou Chen
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Yinghui Huang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Biao Gu
- College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, 421008, PR China
| | - Jiaqi Li
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Cuiyan Wu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Peng Yin
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Youyu Zhang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| | - Haitao Li
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| |
Collapse
|
31
|
Wen L, Shen L. Effect of Surface-Chelated Cu 2+ on Amyloid-β Peptide Fibrillation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:174-181. [PMID: 34932369 DOI: 10.1021/acs.langmuir.1c02322] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Abnormal interactions of copper (Cu) ions with amyloid-β (Aβ) peptides are believed to play an important role in the pathogenesis of Alzheimer's disease (AD). However, there is still debate as to the exact role of Cu ions in Aβ amyloidosis despite extensive studies on Aβ-Cu interactions. Unlike previously reported works, we herein study the effect of surface-chelated Cu2+, rather than the more usual solution-phase dissolved Cu2+, on Aβ aggregation. Through the combination of single molecule fluorescent tracking, atomic force microscopy imaging experiments, and all-atom molecular dynamic simulations, we show that the surface-chelated Cu2+ dynamically interacts with Aβ chains, restricts their 2D-diffusivity on the surface, and retards their fibrillation, while the designated surfaces without Cu2+ facilitate the 2D-diffusivity of Aβ chains for better interpeptide interaction and promote Aβ fibrillation. We offer a microscopic molecular insight into the retardation mechanism of surface-chelated Cu2+ on Aβ fibrillation, suggesting that the surface-bound pools of metal ions are critical in AD progression and drug design.
Collapse
Affiliation(s)
- Lisi Wen
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Lei Shen
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
32
|
Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid. INORGANICS 2022. [DOI: 10.3390/inorganics10010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder that eventually leads the affected patients to die. The appearance of senile plaques in the brains of Alzheimer’s patients is known as a main symptom of this disease. The plaques consist of different components, and according to numerous reports, their main components include beta-amyloid peptide and transition metals such as copper. In this disease, metal dyshomeostasis leads the number of copper ions to simultaneously increase in the plaques and decrease in neurons. Copper ions are essential for proper brain functioning, and one of the possible mechanisms of neuronal death in Alzheimer’s disease is the copper depletion of neurons. However, the reason for the copper depletion is as yet unknown. Based on the available evidence, we suggest two possible reasons: the first is copper released from neurons (along with beta-amyloid peptides), which is deposited outside the neurons, and the second is the uptake of copper ions by activated microglia.
Collapse
|
33
|
Duroux C, Hagège A. CE-ICP-MS to probe Aβ1-42 / copper (II) interactions, a complementary tool to study amyloid aggregation in Alzheimer's Disease. Metallomics 2021; 14:6482862. [PMID: 34951920 DOI: 10.1093/mtomcs/mfab075] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022]
Abstract
Copper (II) ions appear to be involved in the Alzheimer's disease (AD) and seem to influence the aggregation of the amyloid-β1-42 (Aβ1-42) peptide. However, data are not conclusive and still not subject to consensus, copper (II) being suspected to either promote or inhibit aggregation. To address this question, CE-ICP-MS hyphenation was proposed as a complementary tool to follow the distribution of copper in the different oligomeric forms, at different sub-stoichiometries and different incubation times. Results clearly indicated the formation of several negatively charged copper complexes and showed the enhancement of the aggregation rate with copper concentration. Moreover, the variations of copper (II) speciation suggest different aggregation pathway, even for sub-stoichiometric ratios.
Collapse
Affiliation(s)
- C Duroux
- Université de Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 69100 Villeurbanne, France
| | - A Hagège
- Université de Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 69100 Villeurbanne, France
| |
Collapse
|
34
|
Interactions between copper (II) and β-amyloid peptide using capillary electrophoresis-ICP-MS: Kd measurements at the nanogram scale. Anal Bioanal Chem 2021; 414:5347-5355. [PMID: 34812903 DOI: 10.1007/s00216-021-03769-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
Although the interaction between the β-amyloid peptide and copper (II) appears to play an important role in Alzheimer's disease, the affinity constant is still controversial and values are ranging from 107 to 1011 M-1. With the aim of clarifying this point, a complementary method, based on the capillary electrophoresis-ICP-MS hyphenation, was developed and competitive binding experiments were conducted in the presence of nitrilotriacetic acid. The effect of the capillary surface (neutral or positively charged) and nature of the buffer (Tris or Hepes) have been studied. Tris buffer was found to be inappropriate for such determination as it enhances the dissociation of copper (II) complexes, already occurring in the presence of an electric field in capillary electrophoresis. Using Hepes, a value of 1010 M-1 was found for the affinity of the small β-amyloid peptide 1-16 for copper (II), which is in agreement with the values obtained for other proteins involved in neurodegenerative diseases. These constants were also determined in conditions closer to those of biological media (higher ionic strength, presence of carbonates).
Collapse
|
35
|
Yu Y, Zhang Q, Liu G, Deng Y, Kang J, Zhang F, Lu T, Sun L, Qian H. Proteomic analysis of zebrafish brain damage induced by Microcystis aeruginosa bloom. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 795:148865. [PMID: 34246136 DOI: 10.1016/j.scitotenv.2021.148865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
Cyanobacterial blooms constitute a global ecological problem that can seriously threaten human health. One of the most common bloom-forming cyanobacteria in freshwater is Microcystis aeruginosa, whose secretion of toxic substances (microcystins, MCs) have strong liver toxicity and endanger the health of exposed people through contaminated aquatic products and drinking water. However, few studies on the neurotoxicity of M. aeruginosa to zebrafish have simulated the process of an actual cyanobacterial bloom. In this study, we used the zebrafish (Danio rerio) as an effective model organism to study the acute neurotoxicity of M. aeruginosa, and to clarify its principal mechanism of action. A total of 82 upregulated and 26 downregulated proteins were detected by quantitative proteomics analysis in zebrafish brain after exposure to M. aeruginosa. Intriguingly, these proteins with changed expression were related to Synaptic vesicle cycle and terpenoid skeleton biosynthesis pathway, such as ACAT, STX1A, and V-ATPase. The obtained results uniformly indicated that the neurotoxicity of M. aeruginosa seriously damaged the neurotransmitter conduction in the nervous system and brain information storage and transmission of zebrafish and makes it more susceptible to neurological diseases. Our study provides a new perspective on the neurotoxicity risk of cyanobacterial blooms.
Collapse
Affiliation(s)
- Yitian Yu
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qi Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Guangfu Liu
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yu Deng
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jian Kang
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fan Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Tao Lu
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Liwei Sun
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Haifeng Qian
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
36
|
Shardlow E, Khan B, Exley C. Monitoring the early aggregatory behaviour and size of Aβ 1-42 in the absence & presence of metal ions using dynamic light scattering. J Trace Elem Med Biol 2021; 67:126766. [PMID: 33964808 DOI: 10.1016/j.jtemb.2021.126766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM Aβ1-42 is an amyloidogenic peptide found within senile plaques extracted from those who died with a diagnosis of Alzheimer's disease. The potent neurotoxicity of this peptide is related to its propensity to form aggregated conformations in vivo, a process that is influenced by the species and concentration of metal ions present within the local environment. This study examines the impact of different metals upon the early aggregatory behaviour and size of Aβ1-42 under simulated physiological conditions. METHODS The size and aggregatory behaviour of Aβ1-42 in the presence and absence of metal ions was monitored during the initial 30 min of fibril formation in real-time using dynamic light scattering. RESULTS Intensity scattering measurements showed a clear tendency towards aggregation with regards to Aβ1-42 only solutions (10 μM). Both equimolar Al3+ & Cu2+ lowered and stabilised the dimensions of Aβ1-42 aggregates; however, a diminutive but significant increase in size was still observed over a 30-min period. While excess Al3+ continued to supress the size of Aβ1-42, a 10-fold increase in the concentration of Cu2+ accelerated peptide aggregation relative to that observed for equimolar metal but not compared to Aβ1-42 alone. CONCLUSION These results infer that Al3+ ions stabilise and aid in the maintenance of smaller, toxic intermediates while excess Cu2+ facilitates the formation of larger, more inert, amorphous species exceeding 1 μm in size. Furthermore, we propose that metal-induced toxicity of Aβ1-42 is reflective of their ability to preserve smaller oligomeric species in vitro.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Bakhtbilland Khan
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK
| |
Collapse
|
37
|
Fasae KD, Abolaji AO, Faloye TR, Odunsi AY, Oyetayo BO, Enya JI, Rotimi JA, Akinyemi RO, Whitworth AJ, Aschner M. Metallobiology and therapeutic chelation of biometals (copper, zinc and iron) in Alzheimer's disease: Limitations, and current and future perspectives. J Trace Elem Med Biol 2021; 67:126779. [PMID: 34034029 DOI: 10.1016/j.jtemb.2021.126779] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/03/2021] [Accepted: 05/10/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent cause of cognitive impairment and dementia worldwide. The pathobiology of the disease has been studied in the form of several hypotheses, ranging from oxidative stress, amyloid-beta (Aβ) aggregation, accumulation of tau forming neurofibrillary tangles (NFT) through metal dysregulation and homeostasis, dysfunction of the cholinergic system, and to inflammatory and autophagic mechanism. However, none of these hypotheses has led to confirmed diagnostics or approved cure for the disease. OBJECTIVE This review is aimed as a basic and an encyclopedic short course into metals in AD and discusses the advances in chelation strategies and developments adopted in the treatment of the disease. Since there is accumulating evidence of the role of both biometal dyshomeostasis (iron (Fe), copper (Cu), and zinc (Zn)) and metal-amyloid interactions that lead to the pathogenesis of AD, this review focuses on unraveling therapeutic chelation strategies that have been considered in the treatment of the disease, aiming to sequester free and protein-bound metal ions and reducing cerebral metal burden. Promising compounds possessing chemically modified moieties evolving as multi-target ligands used as anti-AD drug candidates are also covered. RESULTS AND CONCLUSION Several multidirectional and multifaceted studies on metal chelation therapeutics show the need for improved synthesis, screening, and analysis of compounds to be able to effectively present chelating anti-AD drugs. Most drug candidates studied have limitations in their physicochemical properties; some enhance redistribution of metal ions, while others indirectly activate signaling pathways in AD. The metal chelation process in vivo still needs to be established and the design of potential anti-AD compounds that bi-functionally sequester metal ions as well as inhibit the Aβ aggregation by competing with the metal ions and reducing metal-induced oxidative damage and neurotoxicity may signal a bright end in chelation-based therapeutics of AD.
Collapse
Affiliation(s)
- Kehinde D Fasae
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Amos O Abolaji
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria.
| | - Tolulope R Faloye
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Atinuke Y Odunsi
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Bolaji O Oyetayo
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Joseph I Enya
- Department of Anatomy, University of Ilorin, Kwara State, Nigeria
| | - Joshua A Rotimi
- Department of Biochemistry and Molecular Biology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Rufus O Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | | | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
38
|
Zhang E, Zhao X, Hu J, Wang R, Fu S, Qin G. Antibacterial metals and alloys for potential biomedical implants. Bioact Mater 2021; 6:2569-2612. [PMID: 33615045 PMCID: PMC7876544 DOI: 10.1016/j.bioactmat.2021.01.030] [Citation(s) in RCA: 147] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/11/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Metals and alloys, including stainless steel, titanium and its alloys, cobalt alloys, and other metals and alloys have been widely used clinically as implant materials, but implant-related infection or inflammation is still one of the main causes of implantation failure. The bacterial infection or inflammation that seriously threatens human health has already become a worldwide complaint. Antibacterial metals and alloys recently have attracted wide attention for their long-term stable antibacterial ability, good mechanical properties and good biocompatibility in vitro and in vivo. In this review, common antibacterial alloying elements, antibacterial standards and testing methods were introduced. Recent developments in the design and manufacturing of antibacterial metal alloys containing various antibacterial agents were described in detail, including antibacterial stainless steel, antibacterial titanium alloy, antibacterial zinc and alloy, antibacterial magnesium and alloy, antibacterial cobalt alloy, and other antibacterial metals and alloys. Researches on the antibacterial properties, mechanical properties, corrosion resistance and biocompatibility of antibacterial metals and alloys have been summarized in detail for the first time. It is hoped that this review could help researchers understand the development of antibacterial alloys in a timely manner, thereby could promote the development of antibacterial metal alloys and the clinical application.
Collapse
Affiliation(s)
- Erlin Zhang
- Key Lab. for Anisotropy and Texture of Materials, Education Ministry of China, School of Materials Science and Engineering, Northeastern University, Shenyang, 150819, China
- Research Center for Metallic Wires, Northeastern University, Shenyang, 110819, China
| | - Xiaotong Zhao
- Key Lab. for Anisotropy and Texture of Materials, Education Ministry of China, School of Materials Science and Engineering, Northeastern University, Shenyang, 150819, China
| | - Jiali Hu
- Key Lab. for Anisotropy and Texture of Materials, Education Ministry of China, School of Materials Science and Engineering, Northeastern University, Shenyang, 150819, China
| | - Ruoxian Wang
- Key Lab. for Anisotropy and Texture of Materials, Education Ministry of China, School of Materials Science and Engineering, Northeastern University, Shenyang, 150819, China
| | - Shan Fu
- Key Lab. for Anisotropy and Texture of Materials, Education Ministry of China, School of Materials Science and Engineering, Northeastern University, Shenyang, 150819, China
| | - Gaowu Qin
- Key Lab. for Anisotropy and Texture of Materials, Education Ministry of China, School of Materials Science and Engineering, Northeastern University, Shenyang, 150819, China
- Research Center for Metallic Wires, Northeastern University, Shenyang, 110819, China
| |
Collapse
|
39
|
Moulahoum H, Ghorbani Zamani F, Timur S, Zihnioglu F. Metal Binding Antimicrobial Peptides in Nanoparticle Bio-functionalization: New Heights in Drug Delivery and Therapy. Probiotics Antimicrob Proteins 2021; 12:48-63. [PMID: 31001788 DOI: 10.1007/s12602-019-09546-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Peptides are considered very important due to the diversity expressed through their amino acid sequence, structure variation, large spectrum, and their essential role in biological systems. Antimicrobial peptides (AMPs) emerged as a potent tool in therapy owing to their antimicrobial properties but also their ability to trespass the membranes, specificity, and low toxicity. They comprise a variety of peptides from which specific amino acid-rich peptides are of interest to the current review due to their features in metal interaction and cell penetration. Histidine-rich peptides such as Histatins belong to the metal binding salivary residing peptides with efficient antibacterial, antifungal, and wound-healing activities. Furthermore, their ability to activate in acidic environment attracted the attention to their potential in therapy. The current review covers the current knowledge about AMPs and critically assess the potential of associating with metal ions both structurally and functionally. This review provides interesting hints for the advantages provided by AMPs and metal ions in biomedicine, making use of their direct properties in brain diseases therapy or in the creation of new bio-functionalized nanoparticles for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Hichem Moulahoum
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey.
| | - Faezeh Ghorbani Zamani
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Suna Timur
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Figen Zihnioglu
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey.
| |
Collapse
|
40
|
The Aggregation Pattern of Aβ
1–40
is Altered by the Presence of
N
‐Truncated Aβ
4–40
and/or Cu
II
in a Similar Way through Ionic Interactions. Chemistry 2021; 27:2798-2809. [DOI: 10.1002/chem.202004484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Indexed: 12/19/2022]
|
41
|
Cioffi F, Adam RHI, Broersen K. Molecular Mechanisms and Genetics of Oxidative Stress in Alzheimer's Disease. J Alzheimers Dis 2020; 72:981-1017. [PMID: 31744008 PMCID: PMC6971833 DOI: 10.3233/jad-190863] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer’s disease is the most common neurodegenerative disorder that can cause dementia in elderly over 60 years of age. One of the disease hallmarks is oxidative stress which interconnects with other processes such as amyloid-β deposition, tau hyperphosphorylation, and tangle formation. This review discusses current thoughts on molecular mechanisms that may relate oxidative stress to Alzheimer’s disease and identifies genetic factors observed from in vitro, in vivo, and clinical studies that may be associated with Alzheimer’s disease-related oxidative stress.
Collapse
Affiliation(s)
- Federica Cioffi
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Kerensa Broersen
- Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
42
|
The Function of Transthyretin Complexes with Metallothionein in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21239003. [PMID: 33256250 PMCID: PMC7730073 DOI: 10.3390/ijms21239003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most frequently diagnosed types of dementia in the elderly. An important pathological feature in AD is the aggregation and deposition of the β-amyloid (Aβ) in extracellular plaques. Transthyretin (TTR) can cleave Aβ, resulting in the formation of short peptides with less activity of amyloid plaques formation, as well as being able to degrade Aβ peptides that have already been aggregated. In the presence of TTR, Aβ aggregation decreases and toxicity of Aβ is abolished. This may prevent amyloidosis but the malfunction of this process leads to the development of AD. In the context of Aβplaque formation in AD, we discuss metallothionein (MT) interaction with TTR, the effects of which depend on the type of MT isoform. In the brains of patients with AD, the loss of MT-3 occurs. On the contrary, MT-1/2 level has been consistently reported to be increased. Through interaction with TTR, MT-2 reduces the ability of TTR to bind to Aβ, while MT-3 causes the opposite effect. It increases TTR-Aβ binding, providing inhibition of Aβ aggregation. The protective effect, assigned to MT-3 against the deposition of Aβ, relies also on this mechanism. Additionally, both Zn7MT-2 and Zn7MT-3, decrease Aβ neurotoxicity in cultured cortical neurons probably because of a metal swap between Zn7MT and Cu(II)Aβ. Understanding the molecular mechanism of metals transfer between MT and other proteins as well as cognition of the significance of TTR interaction with different MT isoforms can help in AD treatment and prevention.
Collapse
|
43
|
Binding of Cu 2+ to Aβ1-29 causes aggregation and toxicity in SH-SY5Y cells. Biochem Biophys Res Commun 2020; 534:617-623. [PMID: 33208229 DOI: 10.1016/j.bbrc.2020.11.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/31/2022]
Abstract
The accumulation and aggregation of amyloid-β (Aβ) are critical factors in the pathogenesis of Alzheimer's disease (AD). Several studies have indicated that metal ions such as Cu2+and Zn2+ play a key role in the formation and stabilization of neurotoxic Aβ aggregates, however the molecular mechanisms underlying Aβ cytotoxicity have not yet been fully elucidated. Previously, we showed that the Aβ-derived fragment peptide (Aβ-FrP), Aβ1-19, altered conformation in the presence of Cu2+, inhibiting its digestion by metalloproteinase-7 (MMP-7). In this study we demonstrated that Aβ1-19 did not form aggregates in the presence of Cu2+. Therefore, we synthesized a new Aβ-FrP, Aβ1-29, which displayed Cu2+-dependent conformational conversion and aggregate formation. Aβ1-29 was cleaved by MMP-7, however this reaction was inhibited in the presence of Cu2+ in a similar way to Aβ1-19. Interestingly, Aβ1-29 showed conformational conversion and aggregate formation in the presence of Zn2+, however this did not confer resistance against MMP-7 cleavage. Moreover, Aβ1-29 induced the apoptotic cell death of neural SH-SY5Y cells in the presence of Cu2+ but not Zn2+. These results suggest that Cu2+, unlike Zn2+, may play an important role in the aggregation mechanism of Aβ and thus in the pathology of AD.
Collapse
|
44
|
Ejaz HW, Wang W, Lang M. Copper Toxicity Links to Pathogenesis of Alzheimer's Disease and Therapeutics Approaches. Int J Mol Sci 2020; 21:E7660. [PMID: 33081348 PMCID: PMC7589751 DOI: 10.3390/ijms21207660] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is an irreversible, age-related progressive neurological disorder, and the most common type of dementia in aged people. Neuropathological lesions of AD are neurofibrillary tangles (NFTs), and senile plaques comprise the accumulated amyloid-beta (Aβ), loaded with metal ions including Cu, Fe, or Zn. Some reports have identified metal dyshomeostasis as a neurotoxic factor of AD, among which Cu ions seem to be a central cationic metal in the formation of plaque and soluble oligomers, and have an essential role in the AD pathology. Cu-Aβ complex catalyzes the generation of reactive oxygen species (ROS) and results in oxidative damage. Several studies have indicated that oxidative stress plays a crucial role in the pathogenesis of AD. The connection of copper levels in AD is still ambiguous, as some researches indicate a Cu deficiency, while others show its higher content in AD, and therefore there is a need to increase and decrease its levels in animal models, respectively, to study which one is the cause. For more than twenty years, many in vitro studies have been devoted to identifying metals' roles in Aβ accumulation, oxidative damage, and neurotoxicity. Towards the end, a short review of the modern therapeutic approach in chelation therapy, with the main focus on Cu ions, is discussed. Despite the lack of strong proofs of clinical advantage so far, the conjecture that using a therapeutic metal chelator is an effective strategy for AD remains popular. However, some recent reports of genetic-regulating copper transporters in AD models have shed light on treating this refractory disease. This review aims to succinctly present a better understanding of Cu ions' current status in several AD features, and some conflicting reports are present herein.
Collapse
Affiliation(s)
- Hafza Wajeeha Ejaz
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China;
| | - Wei Wang
- School of Medical and Health Sciences, Edith Cowan University, Perth WA6027, Australia;
| | - Minglin Lang
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China;
- College of Life Science, Agricultural University of Hebei, Baoding 071000, China
| |
Collapse
|
45
|
Metal- and UV- Catalyzed Oxidation Results in Trapped Amyloid-β Intermediates Revealing that Self-Assembly Is Required for Aβ-Induced Cytotoxicity. iScience 2020; 23:101537. [PMID: 33083713 PMCID: PMC7516296 DOI: 10.1016/j.isci.2020.101537] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/05/2020] [Accepted: 09/03/2020] [Indexed: 02/08/2023] Open
Abstract
Dityrosine (DiY), via the cross-linking of tyrosine residues, is a marker of protein oxidation, which increases with aging. Amyloid-β (Aβ) forms DiY in vitro and DiY-cross-linked Aβ is found in the brains of patients with Alzheimer disease. Metal- or UV- catalyzed oxidation of Aβ42 results in an increase in DiY cross-links. Using DiY as a marker of oxidation, we compare the self-assembly propensity and DiY cross-link formation for a non-assembly competent variant of Aβ42 (vAβ) with wild-type Aβ42. Oxidation results in the formation of trapped wild-type Aβ assemblies with increased DiY cross-links that are unable to elongate further. Assembly-incompetent vAβ and trapped Aβ assemblies are non-toxic to neuroblastoma cells at all stages of self-assembly, in contrast to oligomeric, non-cross-linked Aβ. These findings point to a mechanism of toxicity that necessitates dynamic self-assembly whereby trapped Aβ assemblies and assembly-incompetent variant Aβ are unable to result in cell death. Metal- (Cu2+ H202) or UV- catalyzedoxidation results in dityrosine (DiY) formation Oxidation results in DiY cross-link formation in Aβ and halts further assembly Non-assembling Aβ (trapped Aβ or variant Αβ monomer) is not cytotoxic
Collapse
|
46
|
Rosenberg M, Visnapuu M, Vija H, Kisand V, Kasemets K, Kahru A, Ivask A. Selective antibiofilm properties and biocompatibility of nano-ZnO and nano-ZnO/Ag coated surfaces. Sci Rep 2020; 10:13478. [PMID: 32778787 PMCID: PMC7417576 DOI: 10.1038/s41598-020-70169-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
Spread of pathogenic microbes and antibiotic-resistant bacteria in health-care settings and public spaces is a serious public health challenge. Materials that prevent solid surface colonization or impede touch-transfer of viable microbes could provide means to decrease pathogen transfer from high-touch surfaces in critical applications. ZnO and Ag nanoparticles have shown great potential in antimicrobial applications. Less is known about nano-enabled surfaces. Here we demonstrate that surfaces coated with nano-ZnO or nano-ZnO/Ag composites are not cytotoxic to human keratinocytes and possess species-selective medium-dependent antibiofilm activity against Escherichia coli, Staphylococcus aureus and Candida albicans. Colonization of nano-ZnO and nano-ZnO/Ag surfaces by E. coli and S. aureus was decreased in static oligotrophic conditions (no planktonic growth). Moderate to no effect was observed for bacterial biofilms in growth medium (supporting exponential growth). Inversely, nano-ZnO surfaces enhanced biofilm formation by C. albicans in oligotrophic conditions. However, enhanced C. albicans biofilm formation on nano-ZnO surfaces was effectively counteracted by the addition of Ag. Possible selective enhancement of biofilm formation by the yeast C. albicans on Zn-enabled surfaces should be taken into account in antimicrobial surface development. Our results also indicated the importance of the use of application-appropriate test conditions and exposure medium in antimicrobial surface testing.
Collapse
Affiliation(s)
- M Rosenberg
- Laboratory of Environmental Toxicology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia.
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia.
| | - M Visnapuu
- Institute of Physics, University of Tartu, Tartu, Estonia
| | - H Vija
- Laboratory of Environmental Toxicology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - V Kisand
- Institute of Physics, University of Tartu, Tartu, Estonia
| | - K Kasemets
- Laboratory of Environmental Toxicology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - A Kahru
- Laboratory of Environmental Toxicology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
- Estonian Academy of Sciences, Tallinn, Estonia
| | - A Ivask
- Laboratory of Environmental Toxicology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| |
Collapse
|
47
|
Sasanian N, Bernson D, Horvath I, Wittung-Stafshede P, Esbjörner EK. Redox-Dependent Copper Ion Modulation of Amyloid-β (1-42) Aggregation In Vitro. Biomolecules 2020; 10:E924. [PMID: 32570820 PMCID: PMC7355640 DOI: 10.3390/biom10060924] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
Plaque deposits composed of amyloid-β (Aβ) fibrils are pathological hallmarks of Alzheimer's disease (AD). Although copper ion dyshomeostasis is apparent in AD brains and copper ions are found co-deposited with Aβ peptides in patients' plaques, the molecular effects of copper ion interactions and redox-state dependence on Aβ aggregation remain elusive. By combining biophysical and theoretical approaches, we here show that Cu2+ (oxidized) and Cu+ (reduced) ions have opposite effects on the assembly kinetics of recombinant Aβ(1-42) into amyloid fibrils in vitro. Cu2+ inhibits both the unseeded and seeded aggregation of Aβ(1-42) at pH 8.0. Using mathematical models to fit the kinetic data, we find that Cu2+ prevents fibril elongation. The Cu2+-mediated inhibition of Aβ aggregation shows the largest effect around pH 6.0 but is lost at pH 5.0, which corresponds to the pH in lysosomes. In contrast to Cu2+, Cu+ ion binding mildly catalyzes the Aβ(1-42) aggregation via a mechanism that accelerates primary nucleation, possibly via the formation of Cu+-bridged Aβ(1-42) dimers. Taken together, our study emphasizes redox-dependent copper ion effects on Aβ(1-42) aggregation and thereby provides further knowledge of putative copper-dependent mechanisms resulting in AD.
Collapse
Affiliation(s)
| | | | | | | | - Elin K. Esbjörner
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; (N.S.); (D.B.); (I.H.); (P.W.-S.)
| |
Collapse
|
48
|
Friedemann M, Tõugu V, Palumaa P. Copper(II) partially protects three histidine residues and the N-terminus of amyloid-β peptide from diethyl pyrocarbonate (DEPC) modification. FEBS Open Bio 2020; 10:1072-1081. [PMID: 32255544 PMCID: PMC7262909 DOI: 10.1002/2211-5463.12857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/09/2020] [Accepted: 03/31/2020] [Indexed: 11/07/2022] Open
Abstract
Diethyl pyrocarbonate (DEPC) has been primarily used as a residue‐specific modifying agent to study the role of His residues in peptide/protein and enzyme function; however, its action is not specific, and several other residues can also be modified. In the current study, we monitored the reaction of DEPC with amyloid‐beta (Aβ) peptides and insulin by matrix‐assisted laser desorption/ionization time‐of‐flight mass spectrometry (MALDI‐TOF MS) and determined the modification sites by electrospray ionization quadrupole time‐of‐flight tandem MS (ESI Q‐TOF MS/MS). Our results indicate that five residues in Aβ1–42 are modified in the presence of 30‐fold molar excess of DEPC. After hydroxylamine treatment (which removes modifications from three His residues), two labels remain bound at the peptide N terminus and Lys16. DEPC treatment of Aβ1–42 promotes peptide aggregation, as monitored through the loss of soluble Aβ42 in a semi‐quantitative MALDI‐TOF MS assay. It has been previously proposed that Cu(II) ions protect Aβ1–16 from DEPC modification through binding to His6. We confirmed that Cu(II) ions decrease the stoichiometry of Aβ1–16 modification with the excess of DEPC being lower as compared to the control, which indicates that Cu(II) protects Aβ from DEPC modification. Sequencing of obtained Cu(II)‐protected Aβ1–16 samples showed that Cu(II) does not protect any residues completely, but partially protects all three His residues and the N terminus. Thus, the protection by Cu(II) ions is not related to specific metal binding to a particular residue (e.g. His6), but rather all His residues and the N terminus are involved in binding of Cu(II) ions. These results allow us to elucidate the effect of DEPC modification on amyloidogenity of human Aβ and to speculate about the role of His residues in these processes.
Collapse
Affiliation(s)
- Merlin Friedemann
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| | - Vello Tõugu
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| | - Peep Palumaa
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| |
Collapse
|
49
|
Zhou Y, Liu J, Zheng T, Tian Y. Label-Free SERS Strategy for In Situ Monitoring and Real-Time Imaging of Aβ Aggregation Process in Live Neurons and Brain Tissues. Anal Chem 2020; 92:5910-5920. [PMID: 32227892 DOI: 10.1021/acs.analchem.9b05837] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The aggregation of Aβ has been reported to closely correlate with Alzheimer's disease (AD). However, clear monitoring of the entire aggregation process of Aβ from monomer to fibril has been scarcely reported until now. Herein, we developed a label-free ratiometric surface enhanced Raman spectroscopic (SERS) platform for real-time monitoring of the entire process of Aβ aggregation in neurons and brain tissues. Different gold nanoparticles, generated in situ with Aβ monomer and fibril as templates separately, were served as effective SERS substrates to achieve a high sensitivity with a limit of detection (LOD) down to 70 ± 4 pM and 3.0 ± 0.5 pM for Aβ40 monomer and fibrils, respectively. Besides, the introduction of ratiometric determination of Aβ monomer and fibril (I1244/I1268) realized real-time monitoring of the entire aggregation process of Aβ monomer with high accuracy and selectivity against other proteins and amino acids. The significant analytical performance of the developed platform, together with good biocompatibility, long-term stability, and remarkable spatial resolution, enabled the present SERS platform imaging and real-time monitoring and imaging of Aβ aggregation influenced by different metal ions (Cu2+, Zn2+, and Fe3+) in neurons and brain tissues at the single cell level. Our results suggested that Cu2+ and Zn2+ ion of low concentration (10 μM) promoted fibril formation, while Fe3+ and Zn2+ of high concentration (100 μM) showed inhibition of fibrosis.
Collapse
Affiliation(s)
- Yan Zhou
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| | - Jia Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| | - Tingting Zheng
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China
| |
Collapse
|
50
|
Lu Z, Lu S, Cheng Y, Qin Y, Yang S, Liu X, Fan W, Zheng L, Zhang H. Synthesis of isoniazid-substituted tetraphenylethylene stereoisomers with dramatic differences on aggregate morphologies, optical and mechanocharomic properties. J Photochem Photobiol A Chem 2020. [DOI: 10.1016/j.jphotochem.2020.112357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|