1
|
Xun QQ, Zhang J, Feng L, Ma YY, Li Y, Shi XL. Identification of a novel pyrrolo[2,3- b]pyridine compound as a potent glycogen synthase kinase 3β inhibitor for treating Alzheimer's disease. J Enzyme Inhib Med Chem 2025; 40:2466846. [PMID: 39976249 PMCID: PMC11843656 DOI: 10.1080/14756366.2025.2466846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Herein, a novel pyrrolo[2,3-b]pyridine-based glycogen synthase kinase 3β (GSK-3β) inhibitor, S01, was rationally designed and synthesised to target Alzheimer's disease (AD). S01 inhibited GSK-3β, with an IC50 of 0.35 ± 0.06 nM, and had an acceptable kinase selectivity for 24 structurally similar kinases. Western blotting assays indicated that S01 efficiently increased the expression of p-GSK-3β-Ser9 and decreased p-tau-Ser396 levels in a dose-dependent manner. In vitro cell experiments, S01 showed low cytotoxicity to SH-SY5Y cells, significantly upregulated the expression of β-catenin and neurogenesis-related biomarkers, and effectively promoted the outgrowth of differentiated neuronal neurites. Moreover, S01 substantially ameliorated dyskinesia in AlCl3-induced zebrafish AD models at a concentration of 0.12 μM, which was more potent than Donepezil (8 μM) under identical conditions. Acute toxicity experiments further confirmed the safety of S01 in vivo. Our findings suggested that S01 is a prospective GSK-3β inhibitor and can be tested as a candidate for treating AD.
Collapse
Affiliation(s)
- Qing-Qing Xun
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
- School of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Lei Feng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yu-Ying Ma
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Ying Li
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Xiao-Long Shi
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
2
|
Sohrabi M, Bozorgmehr MR, Momen-Heravi M. Investigating the combined effect of copper, zinc, and iron ions on truncated and full-length Aβ peptides: insights from molecular dynamics simulation. J Biomol Struct Dyn 2025; 43:4165-4173. [PMID: 38189361 DOI: 10.1080/07391102.2024.2301755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
The truncated Aβ1 - 16 peptide containing the metal-binding domain is frequently used in in silico and experimental investigations because it is more soluble and thus more suitable for studies in solution and does not form amyloids. Several spectroscopic studies have shown that the metal binding of Aβ1 - 16 is very similar to that of the full-length Aβ1 - 42. However, since small changes can have a significant impact on aggregation, further experimental and theoretical are needed to elucidate the detailed structures of truncated and full-length Aβ. In this research, the binding of copper ion to the Aβ1 - 16 and Aβ1 - 42 has been studied by molecular dynamics simulation method. To investigate the effect of copper ion on beta-amyloid peptide structure, the simulations were repeated in the copper and zinc ions, copper and iron binary system, and the copper, zinc and iron ions ternary system. The conformation factor was calculated to calculate the binding affinity of copper ion to beta-amyloid peptide residues. The results showed that the initial 16 residues of the beta-amyloid peptide have high binding affinity for copper ions, and histidine 13 and histidine 14 have significantly higher binding affinity for copper ions in all studied systems. Zinc and iron ions were found to reduce the conformational factor of peptide residues in binding to copper ions, and the aggregation tendency was lower in the truncated structure. The SASA results suggest that the side chains of peptide residues are more affected by shortening and the presence of ions.
Collapse
Affiliation(s)
- Mona Sohrabi
- Department of Chemistry, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | | |
Collapse
|
3
|
Valand R, Pandey N, Bellare J, Sivaiah A. A water-soluble glucose-appended quinoline-benzothiazole conjugate as a selective and sensitive receptor for Cu + ions in aqueous media and intracellular bio-imaging in live cells. Analyst 2025; 150:1176-1186. [PMID: 39963841 DOI: 10.1039/d5an00066a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
A water-soluble and biologically benign glucose-appended quinoline-benzothiazole conjugate (L) has been synthesized and characterized using various spectroscopy techniques. The recognition properties of L show selective recognition of Cu+ ions among other biological metal ions including Cu2+ ions in PBS buffer at pH 7.4. L exhibited a switch-on fluorescence enhancement upon the addition of Cu+ with a detection limit of 1.48 × 10-8 M in an aqueous medium. Job's plot confirmed the 1 : 1 binding ratio observed between probe L and Cu+ ions with an association constant (Ka) of 1.46 × 105 M-1. The proposed complex binding mechanism was supported by UV-Vis, fluorescence and ESI-MS. The coordination features of the {L + Cu+} complex were delineated using DFT computational calculations. Furthermore, L was successfully applied for intracellular fluorescence bio-imaging of Cu+ ions in L929 living cells, suggesting that it holds significant potential for Cu+ ion bio-imaging and disease diagnosis.
Collapse
Affiliation(s)
- Ravinkumar Valand
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat-395007, Gujarat, India.
| | - Nidhi Pandey
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai-400076, Mumbai, India
| | - Jayesh Bellare
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai-400076, Mumbai, India
| | - Areti Sivaiah
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat-395007, Gujarat, India.
| |
Collapse
|
4
|
Rana M, Terpstra K, Gutierrez C, Xu K, Arya H, Bhatt TK, Mirica LM, Sharma AK. Evaluation of Anti-Alzheimer's Potential of Azo-Stilbene-Thioflavin-T derived Multifunctional Molecules: Synthesis, Metal and Aβ Species Binding and Cholinesterase Activity. Chemistry 2025; 31:e202402748. [PMID: 39476334 DOI: 10.1002/chem.202402748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024]
Abstract
Inhibition of amyloid β (Aβ) aggregation and cholinesterase activity are two major therapeutic targets for Alzheimer's disease (AD). Multifunctional Molecules (MFMs) specifically designed to address other contributing factors, such as metal ion induced abnormalities, oxidative stress, toxic Aβ aggregates etc. are very much required. Several multifunctional molecules have been developed using different molecular scaffolds. Reported herein is a new series of four MFMs based on ThT, Azo-stilbene and metal ion chelating pockets. The synthesis, characterization, and metal chelation ability for [Cu2+ and Zn2+] are presented herein. Furthermore, we explored their multifunctionality w.r.t. to their (i) recognition of Aβ aggregates and monomeric form, (ii) utility in modulating the aggregation pathways of both metal-free and metal-bound amyloid-β, (iii) ex-vivo staining of amyloid plaques in 5xFAD mice brain sections, (iv) ability to scavenge free radicals and (v) ability to inhibit cholinesterase activity. Molecular docking studies were also performed with Aβ peptides and acetylcholinesterase enzyme to understand the observed inhibitory effect on activity. Overall, the studies presented here establish the multifunctional nature of these molecules and qualify them as promising candidates for furthermore investigation in the quest for finding Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Monika Rana
- Department of Chemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois, 61801, USA
| | - Karna Terpstra
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois, 61801, USA
| | - Citlali Gutierrez
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois, 61801, USA
| | - Kerui Xu
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois, 61801, USA
| | - Hemant Arya
- Department of Biotechnology, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| | - Tarun K Bhatt
- Department of Biotechnology, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| | - Liviu M Mirica
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois, 61801, USA
| | - Anuj K Sharma
- Department of Chemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| |
Collapse
|
5
|
Mahan B, Hu Y, Lahoud E, Nestmeyer M, McCoy-West A, Manestar G, Fowler C, Bush AI, Moynier F. Stable potassium isotope ratios in human blood serum towards biomarker development in Alzheimer's disease. Metallomics 2024; 16:mfae038. [PMID: 39217098 PMCID: PMC11411773 DOI: 10.1093/mtomcs/mfae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
The Alzheimer's disease (AD)-affected brain purges K with concurrently increasing serum K, suggesting brain-blood K transferal. Here, natural stable K isotope ratios-δ41K-of human serum samples were characterized in an AD biomarker pilot study (plus two paired Li-heparin and potassium ethylenediaminetetraacetic acid [K-EDTA] plasma samples). AD serum was found to have a significantly lower mean δ41K relative to controls. To mechanistically explore this change, novel ab initio calculations (density functional theory) of relative K isotope compositions between hydrated K+ and organically bound K were performed, identifying hydrated K+ as isotopically light (lower δ41K) compared to organically bound K. Taken together with literature, serum δ41K and density functional theory results are consistent with efflux of hydrated K+ from the brain to the bloodstream, manifesting a measurable decrease in serum δ41K. These data introduce serum δ41K for further investigation as a minimally invasive AD biomarker, with cost, scalability, and stability advantages over current techniques.
Collapse
Affiliation(s)
- Brandon Mahan
- Melbourne Analytical Geochemistry, School of Geography, Earth and Atmospheric Sciences, University of Melbourne, Melbourne, Australia
- IsoTropics Geochemistry Lab, Earth and Environmental Science, James Cook University, Townsville, Queensland 4814, Australia
| | - Yan Hu
- Institut de Physique du Globe de Paris, Université Paris Cité, CNRS, 75238 Paris, France
- Department of Geoscience, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA
| | - Esther Lahoud
- Institut de Physique du Globe de Paris, Université Paris Cité, CNRS, 75238 Paris, France
| | - Mark Nestmeyer
- IsoTropics Geochemistry Lab, Earth and Environmental Science, James Cook University, Townsville, Queensland 4814, Australia
| | - Alex McCoy-West
- IsoTropics Geochemistry Lab, Earth and Environmental Science, James Cook University, Townsville, Queensland 4814, Australia
| | - Grace Manestar
- Melbourne Analytical Geochemistry, School of Geography, Earth and Atmospheric Sciences, University of Melbourne, Melbourne, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Frédéric Moynier
- Institut de Physique du Globe de Paris, Université Paris Cité, CNRS, 75238 Paris, France
| |
Collapse
|
6
|
Zhang DD. Ironing out the details of ferroptosis. Nat Cell Biol 2024; 26:1386-1393. [PMID: 38429476 DOI: 10.1038/s41556-024-01361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/22/2024] [Indexed: 03/03/2024]
Abstract
Ferroptosis, spurred by excess labile iron and lipid peroxidation, is implicated in various diseases. Advances have been made in comprehending the lipid-peroxidation side of ferroptosis, but the exact role of iron in driving ferroptosis remains unknown. Although iron overload is characterized in multiple disease states, the potential role of ferroptosis within them remains undefined. This overview focuses on the 'ferro' side of ferroptosis, highlighting iron dysregulation in human diseases and potential therapeutic strategies targeting iron regulation and metabolism.
Collapse
Affiliation(s)
- Donna D Zhang
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
| |
Collapse
|
7
|
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 2024; 9:211. [PMID: 39174535 PMCID: PMC11344989 DOI: 10.1038/s41392-024-01911-3] [Citation(s) in RCA: 140] [Impact Index Per Article: 140.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) stands as the predominant form of dementia, presenting significant and escalating global challenges. Its etiology is intricate and diverse, stemming from a combination of factors such as aging, genetics, and environment. Our current understanding of AD pathologies involves various hypotheses, such as the cholinergic, amyloid, tau protein, inflammatory, oxidative stress, metal ion, glutamate excitotoxicity, microbiota-gut-brain axis, and abnormal autophagy. Nonetheless, unraveling the interplay among these pathological aspects and pinpointing the primary initiators of AD require further elucidation and validation. In the past decades, most clinical drugs have been discontinued due to limited effectiveness or adverse effects. Presently, available drugs primarily offer symptomatic relief and often accompanied by undesirable side effects. However, recent approvals of aducanumab (1) and lecanemab (2) by the Food and Drug Administration (FDA) present the potential in disrease-modifying effects. Nevertheless, the long-term efficacy and safety of these drugs need further validation. Consequently, the quest for safer and more effective AD drugs persists as a formidable and pressing task. This review discusses the current understanding of AD pathogenesis, advances in diagnostic biomarkers, the latest updates of clinical trials, and emerging technologies for AD drug development. We highlight recent progress in the discovery of selective inhibitors, dual-target inhibitors, allosteric modulators, covalent inhibitors, proteolysis-targeting chimeras (PROTACs), and protein-protein interaction (PPI) modulators. Our goal is to provide insights into the prospective development and clinical application of novel AD drugs.
Collapse
Affiliation(s)
- Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yinglu Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Yilin Xia
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxian Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
8
|
Zhao R, Sukocheva O, Tse E, Neganova M, Aleksandrova Y, Zheng Y, Gu H, Zhao D, Madhunapantula SV, Zhu X, Liu J, Fan R. Cuproptosis, the novel type of oxidation-induced cell death in thoracic cancers: can it enhance the success of immunotherapy? Cell Commun Signal 2024; 22:379. [PMID: 39068453 PMCID: PMC11282696 DOI: 10.1186/s12964-024-01743-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Copper is an important metal micronutrient, required for the balanced growth and normal physiological functions of human organism. Copper-related toxicity and dysbalanced metabolism were associated with the disruption of intracellular respiration and the development of various diseases, including cancer. Notably, copper-induced cell death was defined as cuproptosis which was also observed in malignant cells, representing an attractive anti-cancer instrument. Excess of intracellular copper leads to the aggregation of lipoylation proteins and toxic stress, ultimately resulting in the activation of cell death. Differential expression of cuproptosis-related genes was detected in normal and malignant tissues. Cuproptosis-related genes were also linked to the regulation of oxidative stress, immune cell responses, and composition of tumor microenvironment. Activation of cuproptosis was associated with increased expression of redox-metabolism-regulating genes, such as ferredoxin 1 (FDX1), lipoic acid synthetase (LIAS), lipoyltransferase 1 (LIPT1), dihydrolipoamide dehydrogenase (DLD), drolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1), and pyruvate dehydrogenase E1 subunit beta (PDHB)). Accordingly, copper-activated network was suggested as an attractive target in cancer therapy. Mechanisms of cuproptosis and regulation of cuproptosis-related genes in different cancers and tumor microenvironment are discussed in this study. The analysis of current findings indicates that therapeutic regulation of copper signaling, and activation of cuproptosis-related targets may provide an effective tool for the improvement of immunotherapy regimens.
Collapse
Affiliation(s)
- Ruiwen Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia.
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yulia Aleksandrova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yufei Zheng
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Deyao Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - SabbaRao V Madhunapantula
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Xiaorong Zhu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruitai Fan
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
9
|
Puello-Silva J, Alí-Torres J. Computational 3D Models of Fe 2+/3+-Aβ 1-42 Complexes Associated with Alzheimer's Disease. J Phys Chem B 2024; 128:7022-7032. [PMID: 39016210 DOI: 10.1021/acs.jpcb.4c01173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The interaction between iron and amyloid-beta (Aβ) peptides has received significant attention in Alzheimer's disease (AD) research due to its potential implications in developing this pathology. However, the coordination preferences of iron and Aβ1-42 have not been thoroughly investigated or remain unknown. This study employs a computational protocol that combines homology modeling techniques with quantum mechanics (DTF-xTB) calculations to build and evaluate several 3D models of Fe2+/3+-Aβ1-42. Our results reveal well-defined complexes for both the metal and peptide moieties, and we discuss the molecular interactions stabilizing these complexes by elucidating the coordinating environments and binding preferences. These proposed models offer valuable insights into the role of iron in Alzheimer's disease (AD) pathology.
Collapse
Affiliation(s)
- Jorge Puello-Silva
- Departamento de Química, Universidad Nacional de Colombia - Sede Bogotá, Bogotá 110111, Colombia
| | - Jorge Alí-Torres
- Departamento de Química, Universidad Nacional de Colombia - Sede Bogotá, Bogotá 110111, Colombia
| |
Collapse
|
10
|
Huang LH, Hsieh YY, Yang FA, Liao WC. DNA-modified Prussian blue nanozymes for enhanced electrochemical biosensing. NANOSCALE 2024; 16:9770-9780. [PMID: 38597919 DOI: 10.1039/d4nr00387j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Prussian blue nanoparticles exhibit the potential to be employed in bioanalytical applications due to their robust stability, peroxidase-like catalytic functionality, straightforward synthesis, and biocompatibility. An efficient approach is presented for the synthesis of nucleic acid-modified Prussian blue nanoparticles (DNA-PBNPs), utilizing nanoparticle porosity to adsorb nucleic acids (polyT). This strategic adsorption leads to the exposure of nucleic acid sequences on the particle surface while retaining catalytic activity. DNA-PBNPs further couple with functional nucleic acid sequences and aptamers through complementary base pairing to act as transducers in biosensors and amplify signal acquisition. Subsequently, we integrated a copper ion-dependent DNAzyme (Cu2+-DNAzyme) and a vascular endothelial growth factor aptamer (VEGF aptamer) onto screen-printed electrodes to serve as recognition elements for analytes. Significantly, our approach leverages DNA-PBNPs as a superior alternative to traditional enzyme-linked antibodies in electrochemical biosensors, thereby enhancing both the efficiency and adaptability of these devices. Our study conclusively demonstrates the application of DNA-PBNPs in two different biosensing paradigms: the sensitive detection of copper ions and vascular endothelial growth factor (VEGF). These results indicate the promising potential of DNA-modified Prussian blue nanoparticles in advancing bioanalytical sensing technologies.
Collapse
Affiliation(s)
- Lin-Hui Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| | - Yu-Yu Hsieh
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| | - Fu-An Yang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| | - Wei-Ching Liao
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
- Medical Device Innovation and Translation Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
11
|
Kastal Z, Balabán A, Vida S, Kállay C, Nagy L, Várnagy K, Sóvágó I. Copper(II), Nickel(II) and Zinc(II) Complexes of Peptide Fragments of Tau Protein. Molecules 2024; 29:2171. [PMID: 38792033 PMCID: PMC11123990 DOI: 10.3390/molecules29102171] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Copper(II), nickel(II) and zinc(II) complexes of various peptide fragments of tau protein were studied by potentiometric and spectroscopic techniques. All peptides contained one histidyl residue and represented the sequences of tau(91-97) (Ac-AQPHTEI-NH2), tau(385-390) (Ac-KTDHGA-NH2) and tau(404-409) (Ac-SPRHLS-NH2). Imidazole-N donors of histidine were the primary metal binding sites for all peptides and all metal ions, but in the case of copper(II) and nickel(II), the deprotonated amide groups were also involved in metal binding by increasing pH. The most stable complexes were formed with copper(II) ions, but the presence of prolyl residues resulted in significant changes in the thermodynamic stability and speciation of the systems. It was also demonstrated that nickel(II) and especially zinc(II) complexes have relatively low thermodynamic stability with these peptides. The copper(II)-catalyzed oxidation of the peptides was also studied. In the presence of H2O2, the fragmentation of peptides was detected in all cases. In the simultaneous presence of H2O2 and ascorbic acid, the fragmentation of the peptide is less preferred, and the formation of 2-oxo-histidine also occurs.
Collapse
Affiliation(s)
- Zsuzsa Kastal
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Z.K.); (C.K.); (K.V.)
| | - Adrienn Balabán
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Z.K.); (C.K.); (K.V.)
| | - Szilvia Vida
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Z.K.); (C.K.); (K.V.)
| | - Csilla Kállay
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Z.K.); (C.K.); (K.V.)
| | - Lajos Nagy
- Department of Applied Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary;
| | - Katalin Várnagy
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Z.K.); (C.K.); (K.V.)
| | - Imre Sóvágó
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Z.K.); (C.K.); (K.V.)
| |
Collapse
|
12
|
Dong Y, Xu T, Yuan L, Wang Y, Yu S, Wang Z, Chen S, Chen C, He W, Stewart T, Zhang W, Yang X. Cerebrospinal fluid efflux through dynamic paracellular pores on venules as a missing piece of the brain drainage system. EXPLORATION (BEIJING, CHINA) 2024; 4:20230029. [PMID: 38855622 PMCID: PMC11022608 DOI: 10.1002/exp.20230029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/31/2023] [Indexed: 06/11/2024]
Abstract
The glymphatic system plays a key role in the clearance of waste from the parenchyma, and its dysfunction has been associated with the pathogenesis of Alzheimer's disease (AD). However, questions remain regarding its complete mechanisms. Here, we report that efflux of cerebrospinal fluid (CSF)/interstitial fluid (ISF) solutes occurs through a triphasic process that cannot be explained by the current model, but rather hints at the possibility of other, previously undiscovered routes from paravenous spaces to the blood. Using real-time, in vivo observation of efflux, a novel drainage pathway was discovered, in which CSF molecules enter the bloodstream directly through dynamically assembled, trumpet-shaped pores (basolateral ϕ<8 μm; apical ϕ < 2 μm) on the walls of brain venules. As Zn2+ could facilitate the brain clearance of macromolecular ISF solutes, Zn2+-induced reconstruction of the tight junctions (TJs) in vascular endothelial cells may participate in pore formation. Thus, an updated model for glymphatic clearance of brain metabolites and potential regulation is postulated. In addition, deficient clearance of Aβ through these asymmetric venule pores was observed in AD model mice, supporting the notion that impaired brain drainage function contributes to Aβ accumulation and pathogenic dilation of the perivascular space in AD.
Collapse
Affiliation(s)
- Yaqiong Dong
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of MedicineQingdao UniversityQingdaoChina
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Ting Xu
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Lan Yuan
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Yahan Wang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Siwang Yu
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Zhi Wang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Shizhu Chen
- The National Institutes of Pharmaceutical R&D Co., Ltd.China Resources Pharmaceutical Group LimitedBeijingChina
| | - Chunhua Chen
- Department of Anatomy and HistologyPeking University Health Science CenterBeijingChina
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry, Coordination Chemistry Institute, School of Chemistry and Chemical EngineeringNanjing UniversityNanjingChina
| | - Tessandra Stewart
- Department of PathologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Weiguang Zhang
- Department of Anatomy and HistologyPeking University Health Science CenterBeijingChina
| | - Xiaoda Yang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
- SATCM Key Laboratory of Compound Drug DetoxificationPeking University Health Science CenterBeijingChina
| |
Collapse
|
13
|
Xing L, Wang Z, Hao Z, Pan P, Yang A, Wang J. Cuproptosis in stroke: focusing on pathogenesis and treatment. Front Mol Neurosci 2024; 17:1349123. [PMID: 38605864 PMCID: PMC11007218 DOI: 10.3389/fnmol.2024.1349123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
Annually, more than 15 million people worldwide suffer from stroke, a condition linked to high mortality and disability rates. This disease significantly affects daily life, impairing everyday functioning, executive function, and cognition. Moreover, stroke severely restricts patients' ability to perform daily activities, diminishing their overall quality of life. Recent scientific studies have identified cuproptosis, a newly discovered form of cell death, as a key factor in stroke development. However, the role of cuproptosis in stroke remains unclear to researchers. Therefore, it is crucial to investigate the mechanisms of cuproptosis in stroke's pathogenesis. This review examines the physiological role of copper, the characteristics and mechanisms of cuproptosis, the differences and similarities between cuproptosis and other cell death types, and the pathophysiology of cuproptosis in stroke, focusing on mitochondrial dysfunction and immune infiltration. Further research is necessary to understand the relationship between previous strokes and cuproptosis and to clarify the mechanisms behind these associations.
Collapse
Affiliation(s)
- Liwei Xing
- The First Clinical Medical School, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Zhifeng Wang
- The First Clinical Medical School, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Zhihui Hao
- The First Clinical Medical School, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Pan Pan
- College of Acupuncture and Massage, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Aiming Yang
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Jian Wang
- The First Clinical Medical School, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
14
|
Zuo Y, Liu HT, Lin LB, Yue RZ, Liu HH, Wang HW, Wang L, Hou RL, Liu WZ, Li CZ, Wang JZ, Li P, Yin YL. A new metal ion chelator attenuates human tau accumulation-induced neurodegeneration and memory deficits in mice. Exp Neurol 2024; 373:114657. [PMID: 38141802 DOI: 10.1016/j.expneurol.2023.114657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023]
Abstract
Neuronal neurofibrillary tangles containing Tau hyperphosphorylation proteins are a typical pathological marker of Alzheimer's disease (AD). The level of tangles in neurons correlates positively with severe dementia. However, how Tau induces cognitive dysfunction is still unknown, which leads to a lack of effective treatments for AD. Metal ions deposition occurs with tangles in AD brain autopsy. Reduced metal ion can improve the pathology of AD. To explore whether abnormally phosphorylated Tau causes metal ion deposition, we overexpressed human full-length Tau (hTau) in the hippocampal CA3 area of mice and primary cultured hippocampal neurons (CPHN) and found that Tau accumulation induced iron deposition and activated calcineurin (CaN), which dephosphorylates glycogen synthase kinase 3 beta (GSK3β), mediating Tau hyperphosphorylation. Simultaneous activation of CaN dephosphorylates cyclic-AMP response binding protein (CREB), leading to synaptic deficits and memory impairment, as shown in our previous study; this seems to be a vicious cycle exacerbating tauopathy. In the current study, we developed a new metal ion chelator that displayed a significant inhibitory effect on Tau phosphorylation and memory impairment by chelating iron ions in vivo and in vitro. These findings provide new insight into the mechanism of memory impairment induced by Tau accumulation and develop a novel potential treatment for tauopathy in AD.
Collapse
Affiliation(s)
- Yue Zuo
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China; School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Hui-Ting Liu
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Lai-Biao Lin
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Rui-Zhu Yue
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Huan-Huan Liu
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China; The Second Affiliated Hospital, Xinxiang Medical University, Xinxiang 453003, China
| | - Hong-Wei Wang
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Lu Wang
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ruan-Ling Hou
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Wei-Zhen Liu
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Chang-Zheng Li
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China; School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Li
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang 453003, China; Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, China.
| | - Ya-Ling Yin
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
15
|
Cheng Z, Han T, Yao J, Wang K, Dong X, Yu F, Huang H, Han M, Liao Q, He S, Lyu W, Li Q. Targeting glycogen synthase kinase-3β for Alzheimer's disease: Recent advances and future Prospects. Eur J Med Chem 2024; 265:116065. [PMID: 38160617 DOI: 10.1016/j.ejmech.2023.116065] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/04/2023] [Accepted: 12/15/2023] [Indexed: 01/03/2024]
Abstract
Senile plaques induced by β-amyloid (Aβ) abnormal aggregation and neurofibrillary tangles (NFT) caused by tau hyperphosphorylation are important pathological manifestations of Alzheimer's disease (AD). Glycogen synthase kinase-3 (GSK-3) is a conserved kinase; one member GSK-3β is highly expressed in the AD brain and involved in the formation of NFT. Hence, pharmacologically inhibiting GSK-3β activity and expression is a good approach to treat AD. As summarized in this article, multiple GSK-3β inhibitors has been comprehensively summarized over recent five years. However, only lithium carbonate and Tideglusib have been studied in clinical trials of AD. Besides ATP-competitive and non-ATP-competitive inhibitors, peptide inhibitors, allosteric inhibitors and other types of inhibitors have gradually attracted more interest. Moreover, considering the close relationship between GSK-3β and other targets involved in cholinergic hypothesis, Aβ aggregation hypothesis, tau hyperphosphorylation hypothesis, oxidative stress hypothesis, neuro-inflammation hypothesis, etc., diverse multifunctional molecules and multi-target directed ligands (MTDLs) have also been disclosed. We hope that these recent advances and critical perspectives will facilitate the discovery of safe and effective GSK-3β inhibitors for AD treatment.
Collapse
Affiliation(s)
- Zimeng Cheng
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Tianyue Han
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Jingtong Yao
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Kaixuan Wang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Xue Dong
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Fan Yu
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - He Huang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Menglin Han
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China
| | - Qinghong Liao
- Shandong Kangqiao Biotechnology Co., Ltd, Qingdao, 266033, Shandong, People's Republic of China
| | - Siyu He
- Guizhou Medical University, Guiyang, 550025, Guizhou, People's Republic of China
| | - Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China
| | - Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, People's Republic of China.
| |
Collapse
|
16
|
Ibrahim IH. Metalloproteins and metalloproteomics in health and disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:123-176. [PMID: 38960472 DOI: 10.1016/bs.apcsb.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Metalloproteins represents more than one third of human proteome, with huge variation in physiological functions and pathological implications, depending on the metal/metals involved and tissue context. Their functions range from catalysis, bioenergetics, redox, to DNA repair, cell proliferation, signaling, transport of vital elements, and immunity. The human metalloproteomic studies revealed that many families of metalloproteins along with individual metalloproteins are dysregulated under several clinical conditions. Also, several sorts of interaction between redox- active or redox- inert metalloproteins are observed in health and disease. Metalloproteins profiling shows distinct alterations in neurodegenerative diseases, cancer, inflammation, infection, diabetes mellitus, among other diseases. This makes metalloproteins -either individually or as families- a promising target for several therapeutic approaches. Inhibitors and activators of metalloenzymes, metal chelators, along with artificial metalloproteins could be versatile in diagnosis and treatment of several diseases, in addition to other biomedical and industrial applications.
Collapse
Affiliation(s)
- Iman Hassan Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| |
Collapse
|
17
|
Du B, Chen K, Wang W, Lei P. Targeting Metals in Alzheimer's Disease: An Update. J Alzheimers Dis 2024; 101:S141-S154. [PMID: 39422951 DOI: 10.3233/jad-240140] [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: 10/19/2024]
Abstract
One pathological feature of Alzheimer's disease (AD) is the dysregulated metal ions, e.g., zinc, copper, and iron in the affected brain regions. The dysregulation of metal homeostasis may cause neurotoxicity and directly addressing these dysregulated metals through metal chelation or mitigating the downstream neurotoxicity stands as a pivotal strategy for AD therapy. This review aims to provide an up-to-date comprehensive overview of the application of metal chelators and drugs targeting metal-related neurotoxicity, such as antioxidants (ferroptotic inhibitors), in the context of AD treatment. It encompasses an exploration of their pharmacological effects, clinical research progress, and potential underlying mechanisms.
Collapse
Affiliation(s)
- Bin Du
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kang Chen
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Weiwei Wang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Elseweidy MM, Mahrous M, Ali SI, Shaheen MA, Younis NN. Pentoxifylline as Add-On Treatment to Donepezil in Copper Sulphate-Induced Alzheimer's Disease-Like Neurodegeneration in Rats. Neurotox Res 2023; 41:546-558. [PMID: 37821782 PMCID: PMC10682165 DOI: 10.1007/s12640-023-00672-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by behavioral, cognitive, and progressive memory impairments. Extensive neuronal loss, extracellular accumulation of insoluble senile amyloid-β (Aβ) plaques, and intracellular neurofibrillary tangles (NFTs) are the major pathological features. The present study aimed to investigate the therapeutic effect of donepezil (DON) and pentoxifylline (PTX) in combination to combat the neurodegenerative disorders (experimental AD) induced by CuSO4 intake in experimental rats. Thirty adult male Wistar rats (140-160 g) were used in this study. AD was first induced in rats by CuSO4 supplement to drinking water (10 mg/L) for 14 weeks. The AD group received no further treatment. Oral treatment with DON (10 mg/kg/day), PTX (100 mg/kg/day), or DON + PTX for the other three groups was started from the 10th week of CuSO4 intake for 4 weeks. Cortex markers like acetylcholine (ACh), acetylcholinesterase (AChE), total antioxidant capacity (TAC), and malondialdehyde (MDA) and hippocampus markers like β-amyloid precursor protein cleaving enzyme 1 (BACE1), phosphorylated Tau (p-tau), Clusterin (CLU), tumor necrosis factor-α (TNF-α), caspase-9 (CAS-9), Bax, and Bcl-2 were measured. The histopathology studies were done by using hematoxylin and eosin and Congo red stains as well as immunohistochemistry for neurofilament. CuSO4 induced adverse histological and biochemical changes. The histological injury in the hippocampus was inhibited following the administration of the DON and PTX. The brain tissue levels of AChE, MDA, BACE1, p-tau, CLU, CAS-9, Bax, and TNF-α were significantly increased, while brain tissue levels of ACh, TAC, and Bcl-2 were significantly decreased in CuSO4-treated rats as compared with the untreated control group. The effects induced by either DON or PTX on most studied parameters were comparable. Combined treatment of DON and PTX induced remarkable results compared with their individual use. However, more clinical and preclinical studies are still required to further confirm and prove the long-term efficacy of such combination.
Collapse
Affiliation(s)
- Mohamed M Elseweidy
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Mohamed Mahrous
- Department of Biochemistry, Faculty of Pharmacy, Port-Said University, Port-Said, 42526, Egypt
| | - Sousou I Ali
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed A Shaheen
- Department of Histology and Cell Biology, Faculty of Human Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Nahla N Younis
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
19
|
Gong L, Sun J, Cong S. Levels of iron and iron-related proteins in Alzheimer's disease: A systematic review and meta-analysis. J Trace Elem Med Biol 2023; 80:127304. [PMID: 37734209 DOI: 10.1016/j.jtemb.2023.127304] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND AND PURPOSE Iron homeostasis disturbance has been suggested to play a role in the pathology of Alzheimer's disease (AD). Systemic iron levels are regulated by iron-related proteins, such as ferritin and transferrin. This meta-analysis was established to evaluate iron and iron-related proteins (ferritin, transferrin, lactoferrin, haptoglobin, hepcidin) in cerebrospinal fluid (CSF) and blood samples of AD patients compared with those in healthy controls (HCs). METHODS Iron and iron-related proteins in Alzheimer's disease was systematically searched within five databases (PubMed, EMBASE, Web of Science, Cochrane, Scopus) up to October 23, 2022. Fifty-four studies (with data for 5105 participants: 2174 AD patients and 2931 HCs) were included in this meta-analysis. This study was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA), applying Stata 14.0 software. RESULTS Decreased iron in blood and increased ferritin in CSF were found in AD patients compared with the levels in HCs. AD patients also exhibited lower lactoferrin in serum. Other variables (iron in CSF, ferritin in blood, transferrin in CSF/blood, haptoglobin in CSF/blood, and hepcidin in blood) did not differ between the groups. CONCLUSION This meta-analysis indicated that iron and iron-related proteins were associated with the risk of AD, suggesting the value of further exploration of iron imbalance in AD using biofluids.
Collapse
Affiliation(s)
- Lin Gong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Jiahui Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Shuyan Cong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
20
|
Marmolejo-Garza A, Krabbendam IE, Luu MDA, Brouwer F, Trombetta-Lima M, Unal O, O'Connor SJ, Majerníková N, Elzinga CRS, Mammucari C, Schmidt M, Madesh M, Boddeke E, Dolga AM. Negative modulation of mitochondrial calcium uniporter complex protects neurons against ferroptosis. Cell Death Dis 2023; 14:772. [PMID: 38007529 PMCID: PMC10676387 DOI: 10.1038/s41419-023-06290-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
Ferroptosis is an iron- and reactive oxygen species (ROS)-dependent form of regulated cell death, that has been implicated in Alzheimer's disease and Parkinson's disease. Inhibition of cystine/glutamate antiporter could lead to mitochondrial fragmentation, mitochondrial calcium ([Ca2+]m) overload, increased mitochondrial ROS production, disruption of the mitochondrial membrane potential (ΔΨm), and ferroptotic cell death. The observation that mitochondrial dysfunction is a characteristic of ferroptosis makes preservation of mitochondrial function a potential therapeutic option for diseases associated with ferroptotic cell death. Mitochondrial calcium levels are controlled via the mitochondrial calcium uniporter (MCU), the main entry point of Ca2+ into the mitochondrial matrix. Therefore, we have hypothesized that negative modulation of MCU complex may confer protection against ferroptosis. Here we evaluated whether the known negative modulators of MCU complex, ruthenium red (RR), its derivative Ru265, mitoxantrone (MX), and MCU-i4 can prevent mitochondrial dysfunction and ferroptotic cell death. These compounds mediated protection in HT22 cells, in human dopaminergic neurons and mouse primary cortical neurons against ferroptotic cell death. Depletion of MICU1, a [Ca2+]m gatekeeper, demonstrated that MICU is protective against ferroptosis. Taken together, our results reveal that negative modulation of MCU complex represents a therapeutic option to prevent degenerative conditions, in which ferroptosis is central to the progression of these pathologies.
Collapse
Affiliation(s)
- Alejandro Marmolejo-Garza
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge E Krabbendam
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Minh Danh Anh Luu
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Famke Brouwer
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Osman Unal
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Shane J O'Connor
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Naďa Majerníková
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Carolina R S Elzinga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy
| | - Martina Schmidt
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Muniswamy Madesh
- Department of Medicine/Cardiology, Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Erik Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
21
|
Madden DJ, Merenstein JL. Quantitative susceptibility mapping of brain iron in healthy aging and cognition. Neuroimage 2023; 282:120401. [PMID: 37802405 PMCID: PMC10797559 DOI: 10.1016/j.neuroimage.2023.120401] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/30/2023] [Indexed: 10/10/2023] Open
Abstract
Quantitative susceptibility mapping (QSM) is a magnetic resonance imaging (MRI) technique that can assess the magnetic properties of cerebral iron in vivo. Although brain iron is necessary for basic neurobiological functions, excess iron content disrupts homeostasis, leads to oxidative stress, and ultimately contributes to neurodegenerative disease. However, some degree of elevated brain iron is present even among healthy older adults. To better understand the topographical pattern of iron accumulation and its relation to cognitive aging, we conducted an integrative review of 47 QSM studies of healthy aging, with a focus on five distinct themes. The first two themes focused on age-related increases in iron accumulation in deep gray matter nuclei versus the cortex. The overall level of iron is higher in deep gray matter nuclei than in cortical regions. Deep gray matter nuclei vary with regard to age-related effects, which are most prominent in the putamen, and age-related deposition of iron is also observed in frontal, temporal, and parietal cortical regions during healthy aging. The third theme focused on the behavioral relevance of iron content and indicated that higher iron in both deep gray matter and cortical regions was related to decline in fluid (speed-dependent) cognition. A handful of multimodal studies, reviewed in the fourth theme, suggest that iron interacts with imaging measures of brain function, white matter degradation, and the accumulation of neuropathologies. The final theme concerning modifiers of brain iron pointed to potential roles of cardiovascular, dietary, and genetic factors. Although QSM is a relatively recent tool for assessing cerebral iron accumulation, it has significant promise for contributing new insights into healthy neurocognitive aging.
Collapse
Affiliation(s)
- David J Madden
- Brain Imaging and Analysis Center, Duke University Medical Center, Box 3918, Durham, NC 27710, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA; Center for Cognitive Neuroscience, Duke University, Durham, NC 27708, USA.
| | - Jenna L Merenstein
- Brain Imaging and Analysis Center, Duke University Medical Center, Box 3918, Durham, NC 27710, USA
| |
Collapse
|
22
|
Abelein A. Metal Binding of Alzheimer's Amyloid-β and Its Effect on Peptide Self-Assembly. Acc Chem Res 2023; 56:2653-2663. [PMID: 37733746 PMCID: PMC10552549 DOI: 10.1021/acs.accounts.3c00370] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 09/23/2023]
Abstract
Metal ions have been identified as key factors modulating the aggregation of amyloid-β peptide (Aβ) implicated in Alzheimer's disease (AD). The presence of elevated levels of metal ions in the amyloid plaques in AD patients supports the notion that the dysfunction of metal homeostasis is connected to the development of AD pathology. Here, recent findings from high- and low-resolution biophysical techniques are put into perspective, providing detailed insights into the molecular structures and dynamics of metal-bound Aβ complexes and the effect of metal ions on the Aβ aggregation process. In particular, the development of theoretical kinetic models deducing different microscopic nucleation events from the macroscopic aggregation behavior has enabled deciphering of the effect of metal ions on specific nucleation processes. In addition to these macroscopic measurements of bulk aggregation to quantify microscopic rates, recent NMR studies have revealed details about the structures and dynamics of metal-Aβ complexes, thereby linking structural events to bulk aggregation. Interestingly, transition-metal ions, such as copper, zinc, and silver ions, form a compact complex with the N-terminal part of monomeric Aβ, respectively, where the metal-bound "folded" state is in dynamic equilibrium with an "unfolded" state. The rates and thermodynamic features of these exchange dynamics have been determined by using NMR relaxation dispersion experiments. Additionally, the application of specifically tailored paramagnetic NMR experiments on the Cu(II)-Aβ complex has been fruitful in obtaining structural constraints within the blind sphere of conventional NMR experiments. This enables the determination of molecular structures of the "folded" Cu(II)-coordinated N-terminal region of Aβ. Furthermore, the discussed transition-metal ions modulate Aβ self-assembly in a concentration-dependent manner, where low metal ion concentrations inhibit Aβ fibril formation, while at high metal ion concentrations other processes occur, resulting in amorphous aggregate formation. Remarkably, the metal-Aβ interaction predominately reduces one specific nucleation step, the fibril-end elongation, whereas primary and surface-catalyzed secondary nucleation mechanisms are less affected. Specific inhibition of fibril-end elongation theoretically predicts an enhanced generation of Aβ oligomers, which is an interesting contribution to understanding metal-Aβ-associated neurotoxic effects. Taken together, the metal binding process creates a metal-bound Aβ complex, which is seemingly inert to aggregation. This process hence efficiently reduces the aggregation-prone peptide pool, which on the macroscopic level is reflected as slower aggregation kinetics. Thus, the specific binding of metals to the Aβ monomer can be linked to the macroscopic inhibitory effect on Aβ bulk aggregation, providing a molecular understanding of the Aβ aggregation mechanism in the presence of metal ions, where the metal ion can be seen as a minimalist agent against Aβ self-assembly. These insights can help to target Aβ aggregation in vivo, where metal ions are key factors modulating the Aβ self-assembly and Aβ-associated neurotoxicity.
Collapse
Affiliation(s)
- Axel Abelein
- Department of Biosciences
and Nutrition, Karolinska Institutet, 141 52 Huddinge, Sweden
| |
Collapse
|
23
|
Cheli VT, Sekhar M, Santiago González DA, Angeliu CG, Denaroso GE, Smith Z, Wang C, Paez PM. The expression of ceruloplasmin in astrocytes is essential for postnatal myelination and myelin maintenance in the adult brain. Glia 2023; 71:2323-2342. [PMID: 37269227 PMCID: PMC10599212 DOI: 10.1002/glia.24424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/05/2023]
Abstract
Ceruloplasmin (Cp) is a ferroxidase enzyme that is essential for cell iron efflux. The absence of this protein in humans and rodents produces progressive neurodegeneration with brain iron accumulation. Astrocytes express high levels of Cp and iron efflux from these cells has been shown to be central for oligodendrocyte maturation and myelination. To explore the role of astrocytic Cp in brain development and aging we generated a specific conditional KO mouse for Cp in astrocytes (Cp cKO). Deletion of Cp in astrocytes during the first postnatal week induced hypomyelination and a significant delay in oligodendrocyte maturation. This abnormal myelin synthesis was exacerbated throughout the first two postnatal months and accompanied by a reduction in oligodendrocyte iron content, as well as an increase in brain oxidative stress. In contrast to young animals, deletion of astrocytic Cp at 8 months of age engendered iron accumulation in several brain areas and neurodegeneration in cortical regions. Aged Cp cKO mice also showed myelin loss and oxidative stress in oligodendrocytes and neurons, and at 18 months of age, developed abnormal behavioral profiles, including deficits in locomotion and short-term memory. In summary, our results demonstrate that iron efflux-mediated by astrocytic Cp-is essential for both early oligodendrocyte maturation and myelin integrity in the mature brain. Additionally, our data suggest that astrocytic Cp activity is central to prevent iron accumulation and iron-induced oxidative stress in the aging CNS.
Collapse
Affiliation(s)
- V T Cheli
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - M Sekhar
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - D A Santiago González
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - C G Angeliu
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - G E Denaroso
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - Z Smith
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - C Wang
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - P M Paez
- Department of Pharmacology and Toxicology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
24
|
Wang L, Wei Y, Sun Z, Jiang LH, Yin Y, Zheng P, Fu Y, Wang H, Li C, Wang JZ. DpdtpA, A Multi-metal Ion Chelator, Attenuates Tau Phosphorylation and Microglial Inflammatory Response via Regulating the PI3K/AKT/GSK-3β Signal Pathways. Neuroscience 2023; 526:196-203. [PMID: 37419407 DOI: 10.1016/j.neuroscience.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023]
Abstract
Tau protein hyperphosphorylation and formation of intracellular neurofibrillary tangles (NFTs) are one of the histopathological hallmarks of Alzheimer's disease (AD) and positively correlated with the severity of AD symptoms. NFTs contain a large number of metal ions that play an important role in regulating tau protein phosphorylation and AD progression. Extracellular tau induces primary phagocytosis of stressed neurons and neuronal loss by activating microglia. Here, we studied the effects of a multi-metal ion chelator, DpdtpA, on tau-induced microglial activation and inflammatory responses and the underlying mechanisms. Treatment with DpdtpA attenuated the increase in the expression of NF-κB and production of inflammatory cytokines, IL-1β, IL-6 and IL-10, in rat microglial cells induced by expression of human tau40 proteins. Treatment with DpdtpA also suppressed tau protein expression and phosphorylation. Moreover, treatment with DpdtpA prevented tau-induced activation of glycogen synthase kinase-3β (GSK-3β) and inhibition of phosphatidylinositol-3-hydroxy kinase (PI3K)/AKT. Collectively, these results show that DpdtpA can attenuate tau phosphorylation and inflammatory responses of microglia by regulating the PI3K/AKT/GSK-3β signal pathways, providing a new option to alleviate neuroinflammation for the treatment of AD.
Collapse
Affiliation(s)
- Lu Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China.
| | - Yingjuan Wei
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Zhenzhou Sun
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang, China
| | - Yaling Yin
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Panpan Zheng
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Yun Fu
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Hongwei Wang
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Changzheng Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Jian-Zhi Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Pyun J, Koay H, Runwal P, Mawal C, Bush AI, Pan Y, Donnelly PS, Short JL, Nicolazzo JA. Cu(ATSM) Increases P-Glycoprotein Expression and Function at the Blood-Brain Barrier in C57BL6/J Mice. Pharmaceutics 2023; 15:2084. [PMID: 37631298 PMCID: PMC10458578 DOI: 10.3390/pharmaceutics15082084] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
P-glycoprotein (P-gp), expressed at the blood-brain barrier (BBB), is critical in preventing brain access to substrate drugs and effluxing amyloid beta (Aβ), a contributor to Alzheimer's disease (AD). Strategies to regulate P-gp expression therefore may impact central nervous system (CNS) drug delivery and brain Aβ levels. As we have demonstrated that the copper complex copper diacetyl bis(4-methyl-3-thiosemicarbazone) (Cu(ATSM)) increases P-gp expression and function in human brain endothelial cells, the present study assessed the impact of Cu(ATSM) on expression and function of P-gp in mouse brain endothelial cells (mBECs) and capillaries in vivo, as well as in peripheral organs. Isolated mBECs treated with Cu(ATSM) (100 nM for 24 h) exhibited a 1.6-fold increase in P-gp expression and a 20% reduction in accumulation of the P-gp substrate rhodamine 123. Oral administration of Cu(ATSM) (30 mg/kg/day) for 28 days led to a 1.5 & 1.3-fold increase in brain microvascular and hepatic expression of P-gp, respectively, and a 20% reduction in BBB transport of [3H]-digoxin. A metallomic analysis showed a 3.5 and 19.9-fold increase in Cu levels in brain microvessels and livers of Cu(ATSM)-treated mice. Our findings demonstrate that Cu(ATSM) increases P-gp expression and function at the BBB in vivo, with implications for CNS drug delivery and clearance of Aβ in AD.
Collapse
Affiliation(s)
- Jae Pyun
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.P.); (P.R.)
| | - HuiJing Koay
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia (P.S.D.)
| | - Pranav Runwal
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.P.); (P.R.)
| | - Celeste Mawal
- Oxidation Biology Lab, Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia; (C.M.); (A.I.B.)
| | - Ashley I. Bush
- Oxidation Biology Lab, Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia; (C.M.); (A.I.B.)
| | - Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.P.); (P.R.)
| | - Paul S. Donnelly
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia (P.S.D.)
| | - Jennifer L. Short
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia;
| | - Joseph A. Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.P.); (P.R.)
| |
Collapse
|
26
|
Güntekin B, Erdal F, Bölükbaş B, Hanoğlu L, Yener G, Duygun R. Alterations of resting-state Gamma frequency characteristics in aging and Alzheimer's disease. Cogn Neurodyn 2023; 17:829-844. [PMID: 37522051 PMCID: PMC10374515 DOI: 10.1007/s11571-022-09873-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/04/2022] [Accepted: 08/13/2022] [Indexed: 11/26/2022] Open
Abstract
Alzheimer's disease (AD) is an important brain disease associated with aging. It involves various functional and structural changes which alter the EEG characteristics. Although numerous studies have found changes in delta, theta, alpha, and beta power, fewer studies have looked at the changes in the resting state EEG gamma activity characteristics in AD. This study aimed to investigate the alterations in the frequency and power values of AD patients' resting-state EEG gamma oscillations compared with healthy elderly and young subjects. We performed Fast Fourier Transform (FFT) on the resting state EEG data from 179 participants, including 59 early stage AD patients, 60 healthy elderly, and 60 healthy young subjects. We averaged FFT performed epochs to investigate the power values in the gamma frequency range (28-48 Hz). We then sorted the peaks of power values in the gamma frequency range, and the average of the identified highest three values was named as the gamma dominant peak frequency. The gamma dominant peak frequency of AD patients (Meyes-opened = 33.4 Hz, Meyes-closed = 32.7 Hz) was lower than healthy elderly (Meyes-opened = 35.5 Hz, Meyes-closed = 35.0 Hz) and healthy young subjects (Meyes-opened = 37.2 Hz, Meyes-closed = 37.0 Hz). These results could be related to AD progression and therefore critical for the recent findings regarding the 40 Hz gamma entrainment because it seems they entrain the gamma frequency of AD towards that of healthy young. Supplementary Information The online version contains supplementary material available at 10.1007/s11571-022-09873-4.
Collapse
Affiliation(s)
- Bahar Güntekin
- Department of Biophysics, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Furkan Erdal
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Neuroscience, Graduate School of Health Science, Istanbul Medipol University, Istanbul, Turkey
- Department of Psychology, Faculty of Arts and Sciences, Marmara University, Istanbul, Turkey
| | - Burcu Bölükbaş
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Neuroscience, Graduate School of Health Science, Istanbul Medipol University, Istanbul, Turkey
| | - Lütfü Hanoğlu
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Neurology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Görsev Yener
- Medical Faculty, Izmir University of Economics, Izmir, Turkey
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Dokuz Eylül University Brain Dynamics Multidisciplinary Research Center, Izmir, Turkey
| | - Rümeysa Duygun
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Neuroscience, Graduate School of Health Science, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
27
|
Park JS, Rehman IU, Choe K, Ahmad R, Lee HJ, Kim MO. A Triterpenoid Lupeol as an Antioxidant and Anti-Neuroinflammatory Agent: Impacts on Oxidative Stress in Alzheimer's Disease. Nutrients 2023; 15:3059. [PMID: 37447385 DOI: 10.3390/nu15133059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease illustrated by neuronal dysfunctions, leading to memory weaknesses and personality changes mostly in the aged population worldwide. The exact cause of AD is unclear, but numerous studies have addressed the involvement of oxidative stress (OS), induced by reactive oxygen species (ROS), to be one of the leading causes in developing AD. OS dysregulates the cellular homeostasis, causing abnormal protein and lipid metabolism. Nutrition plays a pivotal role in modulating the antioxidant system and decreases the neuronal ROS level, thus playing an important therapeutic role in neurodegenerative diseases, especially in AD. Hence, medicinal herbs and their extracts have received global attention as a commercial source of antioxidants Lupeol. Lupeol is a pentacyclic triterpenoid and has many biological functions. It is available in fruits, vegetables, and medicinal plants. It has shown effective antioxidant and anti-inflammatory properties, and higher blood-brain barrier permeability. Also, the binding and inhibitory potentials of Lupeol have been investigated and proved to be effective against certain receptor proteins and enzymes in AD studies by computational molecular docking approaches. Therefore, AD-related research has gained interest in investigating the therapeutic effects of Lupeol. However, despite its beneficial effects in AD, there is still a lack of research in Lupeol. Hence, we compiled in this analysis all preclinical research that looked at Lupeol as an antioxidant and anti-inflammatory agent for AD.
Collapse
Affiliation(s)
- Jun Sung Park
- Division of Life Sciences and Applied Life Science (BK21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Kyonghwan Choe
- Division of Life Sciences and Applied Life Science (BK21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hyeon Jin Lee
- Division of Life Sciences and Applied Life Science (BK21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea
- Alz-Dementia Korea Co., Jinju 52828, Republic of Korea
| |
Collapse
|
28
|
Qian ZM, Li W, Guo Q. Ferroportin1 in the brain. Ageing Res Rev 2023; 88:101961. [PMID: 37236369 DOI: 10.1016/j.arr.2023.101961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/20/2023] [Accepted: 05/20/2023] [Indexed: 05/28/2023]
Abstract
Despite years of research, it remains unclear why certain brain regions of patients with neurodegenerative diseases (NDs) have abnormally high levels of iron, although it has long been suggested that disrupted expression of iron-metabolizing proteins due to genetic or non-genetic factors is responsible for the enhancement in brain iron contents. In addition to the increased expression of cell-iron importers lactoferrin (lactotransferrin) receptor (LfR) in Parkinson's disease (PD) and melanotransferrin (p97) in Alzheimer's disease (AD), some investigations have suggested that cell-iron exporter ferroportin 1 (Fpn1) may be also associated with the elevated iron observed in the brain. The decreased expression of Fpn1 and the resulting decrease in the amount of iron excreted from brain cells has been thought to be able to enhance iron levels in the brain in AD, PD and other NDs. Cumulative results also suggest that the reduction of Fpn1 can be induced by hepcidin-dependent and -independent pathways. In this article, we discuss the current understanding of Fpn1 expression in the brain and cell lines of rats, mice and humans, with emphasis on the potential involvement of reduced Fpn1 in brain iron enhancement in patients with AD, PD and other NDs.
Collapse
Affiliation(s)
- Zhong-Ming Qian
- Department of Neurology, Affiliated Hospital of Nantong University, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu China 226019.
| | - Wei Li
- Department of Neurology, Affiliated Hospital of Nantong University, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu China 226019
| | - Qian Guo
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, 881 Yonghe Road, Nantong, Jiangsu 226001, China; Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China.
| |
Collapse
|
29
|
Ma J, Guo Q, Shen MQ, Li W, Zhong QX, Qian ZM. Apolipoprotein E is required for brain iron homeostasis in mice. Redox Biol 2023; 64:102779. [PMID: 37339558 PMCID: PMC10363452 DOI: 10.1016/j.redox.2023.102779] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Apolipoprotein E deficiency (ApoE-/-) increases progressively iron in the liver, spleen and aortic tissues with age in mice. However, it is unknown whether ApoE affects brain iron. METHODS We investigated iron contents, expression of transferrin receptor 1 (TfR1), ferroportin 1 (Fpn1), iron regulatory proteins (IRPs), aconitase, hepcidin, Aβ42, MAP2, reactive oxygen species (ROS), cytokines and glutathione peroxidase 4 (Gpx4) in the brain of ApoE-/- mice. RESULTS We demonstrated that ApoE-/- induced a significant increase in iron, TfR1 and IRPs and a reduction in Fpn1, aconitase and hepcidin in the hippocampus and basal ganglia. We also showed that replenishment of ApoE absent partly reversed the iron-related phenotype in ApoE-/- mice at 24-months old. In addition, ApoE-/- induced a significant increase in Aβ42, MDA, 8-isoprostane, IL-1β, IL-6, and TNFα and a reduction in MAP2 and Gpx4 in hippocampus, basal ganglia and/or cortex of mice at 24-months old. CONCLUSIONS Our findings implied that ApoE is required for brain iron homeostasis and ApoE-/--induced increase in brain iron is due to the increased IRP/TfR1-mediated cell-iron uptake as well as the reduced IRP/Fpn1 associated cell-iron export and suggested that ApoE-/- induced neuronal injury resulted mainly from the increased iron and subsequently ROS, inflammation and ferroptosis.
Collapse
Affiliation(s)
- Juan Ma
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China; Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China.
| | - Qian Guo
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, 881 Yonghe Road, Nantong, Jiangsu, 226001, China; Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
| | - Meng-Qi Shen
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China.
| | - Wei Li
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China.
| | - Qi-Xin Zhong
- Department of Cardiovascular Medicine, Shenzhen Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518034, China.
| | - Zhong-Ming Qian
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
30
|
Vicente-Zurdo D, Brunetti L, Piemontese L, Guedes B, Cardoso SM, Chavarria D, Borges F, Madrid Y, Chaves S, Santos MA. Rivastigmine-Benzimidazole Hybrids as Promising Multitarget Metal-Modulating Compounds for Potential Treatment of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24098312. [PMID: 37176018 PMCID: PMC10179505 DOI: 10.3390/ijms24098312] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
With the goal of combating the multi-faceted Alzheimer's disease (AD), a series of Rivastigmine-Benzimidazole (RIV-BIM) hybrids was recently reported by us as multitarget-directed ligands, thanks to their capacity to tackle important hallmarks of AD. In particular, they exhibited antioxidant activity, acted as cholinesterase inhibitors, and inhibited amyloid-β (Aβ) aggregation. Herein, we moved forward in this project, studying their ability to chelate redox-active biometal ions, Cu(II) and Fe(III), with widely recognized roles in the generation of oxidative reactive species and in protein misfolding and aggregation in both AD and Parkinson's disease (PD). Although Cu(II) chelation showed higher efficiency for the positional isomers of series 5 than those of series 4 of the hybrids, the Aβ-aggregation inhibition appears more dependent on their capacity for fibril intercalation than on copper chelation. Since monoamine oxidases (MAOs) are also important targets for the treatment of AD and PD, the capacity of these hybrids to inhibit MAO-A and MAO-B was evaluated, and they showed higher activity and selectivity for MAO-A. The rationalization of the experimental evaluations (metal chelation and MAO inhibition) was supported by computational molecular modeling studies. Finally, some compounds showed also neuroprotective effects in human neuroblastoma (SH-SY5Y cells) upon treatment with 1-methyl-4-phenylpyridinium (MPP+), a neurotoxic metabolite of a Parkinsonian-inducing agent.
Collapse
Affiliation(s)
- David Vicente-Zurdo
- Centro de Química Estrutural, Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| | - Leonardo Brunetti
- Centro de Química Estrutural, Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125 Bari, Italy
| | - Luca Piemontese
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125 Bari, Italy
| | - Beatriz Guedes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Sandra M Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3000-370 Coimbra, Portugal
- FMUC-Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Daniel Chavarria
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Yolanda Madrid
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| | - Sílvia Chaves
- Centro de Química Estrutural, Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
| | - M Amélia Santos
- Centro de Química Estrutural, Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
| |
Collapse
|
31
|
Wang M, Tang G, Zhou C, Guo H, Hu Z, Hu Q, Li G. Revisiting the intersection of microglial activation and neuroinflammation in Alzheimer's disease from the perspective of ferroptosis. Chem Biol Interact 2023; 375:110387. [PMID: 36758888 DOI: 10.1016/j.cbi.2023.110387] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/12/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by chronic neuroinflammation with amyloid beta-protein deposition and hyperphosphorylated tau protein. The typical clinical manifestation of AD is progressive memory impairment, and AD is considered a multifactorial disease with various etiologies (genetic factors, aging, lifestyle, etc.) and complicated pathophysiological processes. Previous research identified that neuroinflammation and typical microglial activation are significant mechanisms underlying AD, resulting in dysfunction of the nervous system and progression of the disease. Ferroptosis is a novel modality involved in this process. As an iron-dependent form of cell death, ferroptosis, characterized by iron accumulation, lipid peroxidation, and irreversible plasma membrane disruption, promotes AD by accelerating neuronal dysfunction and abnormal microglial activation. In this case, disturbances in brain iron homeostasis and neuronal ferroptosis aggravate neuroinflammation and lead to the abnormal activation of microglia. Abnormally activated microglia release various pro-inflammatory factors that aggravate the dysregulation of iron homeostasis and neuroinflammation, forming a vicious cycle. In this review, we first introduce ferroptosis, microglia, AD, and their relationship. Second, we discuss the nonnegligible role of ferroptosis in the abnormal microglial activation involved in the chronic neuroinflammation of AD to provide new ideas for the identification of potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Miaomiao Wang
- Queen Mary School, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Gan Tang
- Queen Mary School, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Congfa Zhou
- Department of Anatomy, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Hongmin Guo
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Zihui Hu
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Qixing Hu
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Guilin Li
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
32
|
Babić Leko M, Langer Horvat L, Španić Popovački E, Zubčić K, Hof PR, Šimić G. Metals in Alzheimer's Disease. Biomedicines 2023; 11:1161. [PMID: 37189779 PMCID: PMC10136077 DOI: 10.3390/biomedicines11041161] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
The role of metals in the pathogenesis of Alzheimer's disease (AD) is still debated. Although previous research has linked changes in essential metal homeostasis and exposure to environmental heavy metals to the pathogenesis of AD, more research is needed to determine the relationship between metals and AD. In this review, we included human studies that (1) compared the metal concentrations between AD patients and healthy controls, (2) correlated concentrations of AD cerebrospinal fluid (CSF) biomarkers with metal concentrations, and (3) used Mendelian randomization (MR) to assess the potential metal contributions to AD risk. Although many studies have examined various metals in dementia patients, understanding the dynamics of metals in these patients remains difficult due to considerable inconsistencies among the results of individual studies. The most consistent findings were for Zn and Cu, with most studies observing a decrease in Zn levels and an increase in Cu levels in AD patients. However, several studies found no such relation. Because few studies have compared metal levels with biomarker levels in the CSF of AD patients, more research of this type is required. Given that MR is revolutionizing epidemiologic research, additional MR studies that include participants from diverse ethnic backgrounds to assess the causal relationship between metals and AD risk are critical.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Ena Španić Popovački
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Klara Zubčić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute and Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
33
|
Sharma P, Singh M. An ongoing journey of chalcone analogues as single and multi-target ligands in the field of Alzheimer's disease: A review with structural aspects. Life Sci 2023; 320:121568. [PMID: 36925061 DOI: 10.1016/j.lfs.2023.121568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Alzheimer's disease (AD) is a chronic and irreversible neurodegenerative disorder with progressive dementia and cognitive impairment. AD poses severe health challenge in elderly people and become one of the leading causes of death worldwide. It possesses complex pathophysiology with several hypotheses (cholinergic hypothesis, amyloid hypothesis, tau hypothesis, oxidative stress, mitochondrial dysfunction etc.). Several attempts have been made for the management of multifactorial AD. Acetylcholinesterase is the only target has been widely explored in the management of AD to the date. The current review set forth the chalcone based natural, semi-synthetic and synthetic compounds in the search of potential anti-Alzheimer's agents. The main highlights of current review emphasizes on chalcone target different enzymes and pathways like Acetylcholinesterase, β-secretase (BACE1), tau proteins, MAO, free radicals, Advanced glycation end Products (AGEs) etc. and their structure activity relationships contributing in the inhibition of above mentioned various targets of AD.
Collapse
Affiliation(s)
- Pratibha Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
34
|
Yu J, Wu D, Zhao Y, Guo L, Liu P. Study on multi-target effects of PIMPC on Aβ/Cu 2+-induced Alzheimer's disease model of rats. Brain Res 2023; 1802:148226. [PMID: 36586663 DOI: 10.1016/j.brainres.2022.148226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/22/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
Glycogen synthase kinase-3 (GSK-3), a key role in the pathogenesis of Alzheimer's disease (AD), has been linked with the formation of β-amyloid (Aβ), tubulin-associated unit (tau) protein phosphorylation and apoptosis. Moreover, the excessive presence of elements such as copper (Cu) can promote Aβ aggregation and increase the risk of AD. Combined with the role of GSK-3 and metal elements in AD, a metal-chelating imine GSK-3 inhibitor N-(4-{[(2-amino-5-phenylpyridin-3-ylidene)imino]methyl}pyridin-2-yl)cyclopropanecarboxamide (PIMPC) was designed and synthesized. In our study, Aβ/Cu2+-induced AD rat model was established and treated with PIMPC. The results indicated that PIMPC can not only down-regulate the high expression levels of Aβ, tau and p-tau proteins of the AD rats, but also chelate Cu and aluminum (Al) elements in the brain. In addition, PIMPC may play an anti-apoptotic effect by down-regulating the high expression of cleaved Caspase-3 protein, and it can modulate ATPase and nitric oxide synthase (NOS) levels, oxidative stress and neurotransmitter disturbance. In summary, PIMPC acts on multiple targets to relieve the learning and memory impairment of AD rats induced by Aβ/Cu2+.
Collapse
Affiliation(s)
- Jiasi Yu
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Dan Wu
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yanan Zhao
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Linli Guo
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ping Liu
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
35
|
Berntsson E, Vosough F, Svantesson T, Pansieri J, Iashchishyn IA, Ostojić L, Dong X, Paul S, Jarvet J, Roos PM, Barth A, Morozova-Roche LA, Gräslund A, Wärmländer SKTS. Residue-specific binding of Ni(II) ions influences the structure and aggregation of amyloid beta (Aβ) peptides. Sci Rep 2023; 13:3341. [PMID: 36849796 PMCID: PMC9971182 DOI: 10.1038/s41598-023-29901-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. AD brains display deposits of insoluble amyloid plaques consisting mainly of aggregated amyloid-β (Aβ) peptides, and Aβ oligomers are likely a toxic species in AD pathology. AD patients display altered metal homeostasis, and AD plaques show elevated concentrations of metals such as Cu, Fe, and Zn. Yet, the metal chemistry in AD pathology remains unclear. Ni(II) ions are known to interact with Aβ peptides, but the nature and effects of such interactions are unknown. Here, we use numerous biophysical methods-mainly spectroscopy and imaging techniques-to characterize Aβ/Ni(II) interactions in vitro, for different Aβ variants: Aβ(1-40), Aβ(1-40)(H6A, H13A, H14A), Aβ(4-40), and Aβ(1-42). We show for the first time that Ni(II) ions display specific binding to the N-terminal segment of full-length Aβ monomers. Equimolar amounts of Ni(II) ions retard Aβ aggregation and direct it towards non-structured aggregates. The His6, His13, and His14 residues are implicated as binding ligands, and the Ni(II)·Aβ binding affinity is in the low µM range. The redox-active Ni(II) ions induce formation of dityrosine cross-links via redox chemistry, thereby creating covalent Aβ dimers. In aqueous buffer Ni(II) ions promote formation of beta sheet structure in Aβ monomers, while in a membrane-mimicking environment (SDS micelles) coil-coil helix interactions appear to be induced. For SDS-stabilized Aβ oligomers, Ni(II) ions direct the oligomers towards larger sizes and more diverse (heterogeneous) populations. All of these structural rearrangements may be relevant for the Aβ aggregation processes that are involved in AD brain pathology.
Collapse
Affiliation(s)
- Elina Berntsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden.
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia.
| | - Faraz Vosough
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Teodor Svantesson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Jonathan Pansieri
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Igor A Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Lucija Ostojić
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Xiaolin Dong
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Suman Paul
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
- The National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Per M Roos
- Institute of Environmental Medicine, Karolinska Institutet, Nobels Väg 13, 171 77, Stockholm, Sweden
- Department of Clinical Physiology, Capio St. Göran Hospital, St. Göransplan 1, 112 19, Stockholm, Sweden
| | - Andreas Barth
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | | | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | | |
Collapse
|
36
|
A Bifunctional Fluorescence Probe Based on AIE-ICT Strategy for Visual Detection of Cu 2+/Co 2+ in Complex Matrix. Molecules 2023; 28:molecules28052059. [PMID: 36903303 PMCID: PMC10003869 DOI: 10.3390/molecules28052059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
A novel fluorescence chemical sensor-based probe 1-{[(E)-(2-aminophenyl)azanylidene]methyl}naphthalen-2-ol (AMN) was designed and synthesized, which performed a "naked eye" detection ability toward Cu2+ and Co2+ based on aggregation-induced emission (AIE) fluorescence strategy. It has sensitive detection ability for Cu2+ and Co2+. In addition, the color changed from yellow-green to orange under the sunlight, realizing the rapid identification of Cu2+/Co2+, which has the potential of on-site visual detection under the "naked eye". Moreover, different "on" and "off" fluorescence expressions were exhibited under excessive glutathione (GSH) in AMN-Cu2+ and AMN-Co2+ systems, which could be employed to distinguish Cu2+ from Co2+. The detection limits for Cu2+ and Co2+ were measured to be 8.29 × 10-8 M and 9.13 × 10-8 M, respectively. The binding mode of AMN was calculated to be 2:1 by Jobs' plot method analysis. Ultimately, the new fluorescence sensor was applied to detect Cu2+ and Co2+ in real samples (tap water, river water, and yellow croaker), and the results were satisfying. Therefore, this high-efficiency bifunctional chemical sensor platform based on "on-off" fluorescence detection will provide significant guidance for the advance development of single-molecule sensors for multi-ion detection.
Collapse
|
37
|
Andrikopoulos N, Li Y, Nandakumar A, Quinn JF, Davis TP, Ding F, Saikia N, Ke PC. Zinc-Epigallocatechin-3-gallate Network-Coated Nanocomposites against the Pathogenesis of Amyloid-Beta. ACS APPLIED MATERIALS & INTERFACES 2023; 15:7777-7792. [PMID: 36724494 PMCID: PMC10037301 DOI: 10.1021/acsami.2c20334] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The aggregation of amyloid beta (Aβ) is a hallmark of Alzheimer's disease (AD), a major cause of dementia and an unmet challenge in modern medicine. In this study, we constructed a biocompatible metal-phenolic network (MPN) comprising a polyphenol epigallocatechin gallate (EGCG) scaffold coordinated by physiological Zn(II). Upon adsorption onto gold nanoparticles, the MPN@AuNP nanoconstruct elicited a remarkable potency against the amyloid aggregation and toxicity of Aβ in vitro. The superior performance of MPN@AuNP over EGCG@AuNP was attributed to the porosity and hence larger surface area of the MPN in comparison with that of EGCG alone. The atomic detail of Zn(II)-EGCG coordination was unraveled by density functional theory calculations and the structure and dynamics of Aβ aggregation modulated by the MPN were further examined by discrete molecular dynamics simulations. As MPN@AuNP also displayed a robust capacity to cross a blood-brain barrier model through the paracellular pathway, and given the EGCG's function as an anti-amyloidosis and antioxidation agent, this MPN-based strategy may find application in regulating the broad AD pathology beyond protein aggregation inhibition.
Collapse
Affiliation(s)
- Nicholas Andrikopoulos
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yuhuan Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Aparna Nandakumar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - John F. Quinn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Department of Chemical and Biological Engineering, Faculty of Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Thomas P. Davis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Nabanita Saikia
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| |
Collapse
|
38
|
Puentes-Díaz N, Chaparro D, Morales-Morales D, Flores-Gaspar A, Alí-Torres J. Role of Metal Cations of Copper, Iron, and Aluminum and Multifunctional Ligands in Alzheimer's Disease: Experimental and Computational Insights. ACS OMEGA 2023; 8:4508-4526. [PMID: 36777601 PMCID: PMC9909689 DOI: 10.1021/acsomega.2c06939] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/30/2022] [Indexed: 05/15/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, affecting millions of people around the world. Even though the causes of AD are not completely understood due to its multifactorial nature, some neuropathological hallmarks of its development have been related to the high concentration of some metal cations. These roles include the participation of these metal cations in the production of reactive oxygen species, which have been involved in neuronal damage. In order to avoid the increment in the oxidative stress, multifunctional ligands used to coordinate these metal cations have been proposed as a possible treatment to AD. In this review, we present the recent advances in experimental and computational works aiming to understand the role of two redox active and essential transition-metal cations (Cu and Fe) and one nonbiological metal (Al) and the recent proposals on the development of multifunctional ligands to stop or revert the damaging effects promoted by these metal cations.
Collapse
Affiliation(s)
- Nicolás Puentes-Díaz
- Departamento
de Química, Universidad Nacional
de Colombia−Sede Bogotá, Bogotá 11301, Colombia
| | - Diego Chaparro
- Departamento
de Química, Universidad Nacional
de Colombia−Sede Bogotá, Bogotá 11301, Colombia
- Departamento
de Química, Universidad Militar Nueva
Granada, Cajicá 250240, Colombia
| | - David Morales-Morales
- Instituto
de Química, Universidad Nacional Autónoma de México,
Circuito Exterior, Ciudad Universitaria, Ciudad de México 04510, México
| | - Areli Flores-Gaspar
- Departamento
de Química, Universidad Militar Nueva
Granada, Cajicá 250240, Colombia
- Areli Flores-Gaspar − Departamento de Química,
Universidad Militar Nueva
Granada, Cajicá, 250247, Colombia.
| | - Jorge Alí-Torres
- Departamento
de Química, Universidad Nacional
de Colombia−Sede Bogotá, Bogotá 11301, Colombia
- Jorge Alí-Torres − Departamento de Química, Universidad Nacional de
Colombia, Sede Bogotá,11301, Bogotá, Colombia.
| |
Collapse
|
39
|
Zhang E, Dai F, Chen T, Liu S, Xiao C, Shen X. Diagnostic models and predictive drugs associated with cuproptosis hub genes in Alzheimer's disease. Front Neurol 2023; 13:1064639. [PMID: 36776574 PMCID: PMC9909238 DOI: 10.3389/fneur.2022.1064639] [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: 10/08/2022] [Accepted: 12/29/2022] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease, and its underlying genes and treatments are unclear. Abnormalities in copper metabolism can prevent the clearance of β-amyloid peptides and promote the progression of AD pathogenesis. Therefore, the present study used a bioinformatics approach to perform an integrated analysis of the hub gene based on cuproptosis that can influence the diagnosis and treatment of AD. The gene expression profiles were obtained from the Gene Expression Omnibus database, including non-demented (ND) and AD samples. A total of 2,977 cuproptosis genes were retrieved from published articles. The seven hub genes associated with cuproptosis and AD were obtained from the differentially expressed genes and WGCNA in brain tissue from GSE33000. The GO analysis demonstrated that these genes were involved in phosphoribosyl pyrophosphate, lipid, and glucose metabolism. By stepwise regression and logistic regression analysis, we screened four of the seven cuproptosis genes to construct a diagnostic model for AD, which was validated by GES15222, GS48350, and GSE5281. In addition, immune cell infiltration of samples was investigated for correlation with these hub genes. We identified six drugs targeting these seven cuproptosis genes in DrugBank. Hence, these cuproptosis gene signatures may be an important prognostic indicator for AD and may offer new insights into treatment options.
Collapse
Affiliation(s)
- Erdong Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Guiyang, Guizhou, China
| | - Fengqiu Dai
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Tingting Chen
- Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Shanhui Liu
- Key Laboratory of Urological Diseases in Gansu Province, Gansu Nephro-Urological Clinical Center, Institute of Urology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Chaolun Xiao
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiangchun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
40
|
Xie D, Deng T, Zhai Z, Sun T, Xu Y. The cellular model for Alzheimer's disease research: PC12 cells. Front Mol Neurosci 2023; 15:1016559. [PMID: 36683856 PMCID: PMC9846650 DOI: 10.3389/fnmol.2022.1016559] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD) is a common age-related neurodegenerative disease characterized by progressive cognitive decline and irreversible memory impairment. Currently, several studies have failed to fully elucidate AD's cellular and molecular mechanisms. For this purpose, research on related cellular models may propose potential predictive models for the drug development of AD. Therefore, many cells characterized by neuronal properties are widely used to mimic the pathological process of AD, such as PC12, SH-SY5Y, and N2a, especially the PC12 pheochromocytoma cell line. Thus, this review covers the most systematic essay that used PC12 cells to study AD. We depict the cellular source, culture condition, differentiation methods, transfection methods, drugs inducing AD, general approaches (evaluation methods and metrics), and in vitro cellular models used in parallel with PC12 cells.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
41
|
Djafarou S, Amine Khodja I, Boulebd H. Computational design of new tacrine analogs: an in silico prediction of their cholinesterase inhibitory, antioxidant, and hepatotoxic activities. J Biomol Struct Dyn 2023; 41:91-105. [PMID: 34825629 DOI: 10.1080/07391102.2021.2004232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tacrine, the first drug approved for the treatment of Alzheimer's disease (AD), is a non-competitive cholinesterase inhibitor withdrawn due to its acute hepatotoxicity. However, new non-hepatotoxic forms of tacrine have been actively researched. Moreover, several recent reports have shown that oxidative stress is the cause of damage and plays a role in the pathogenesis of several neurodegenerative diseases including AD. The aim of the present study is the design of new easily synthesized tacrine analogs with less hepatotoxicity and potent antioxidant activity. In this context, a library of 34 novel tacrine analogs bearing an antioxidant fragment was designed and evaluated for its hepatotoxicity as well as anticholinesterase and antioxidant activities using computational methods. As a result, six new tacrine analogs have been proposed as potential inhibitors of cholinesterase with antioxidant activity and low or no hepatotoxicity. Furthermore, ADME calculations suggest that these compounds are promising oral drug candidates. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Selsabil Djafarou
- Laboratory of Synthesis of Molecules with Biological Interest, University of Frères Mentouri Constantine 1, Constantine, Algeria
| | - Imene Amine Khodja
- Laboratory of Synthesis of Molecules with Biological Interest, University of Frères Mentouri Constantine 1, Constantine, Algeria
| | - Houssem Boulebd
- Laboratory of Synthesis of Molecules with Biological Interest, University of Frères Mentouri Constantine 1, Constantine, Algeria
| |
Collapse
|
42
|
Babić Leko M, Mihelčić M, Jurasović J, Nikolac Perković M, Španić E, Sekovanić A, Orct T, Zubčić K, Langer Horvat L, Pleić N, Kiđemet-Piskač S, Vogrinc Ž, Pivac N, Diana A, Borovečki F, Hof PR, Šimić G. Heavy Metals and Essential Metals Are Associated with Cerebrospinal Fluid Biomarkers of Alzheimer's Disease. Int J Mol Sci 2022; 24:467. [PMID: 36613911 PMCID: PMC9820819 DOI: 10.3390/ijms24010467] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Various metals have been associated with the pathogenesis of Alzheimer's disease (AD), principally heavy metals that are environmental pollutants (such as As, Cd, Hg, and Pb) and essential metals whose homeostasis is disturbed in AD (such as Cu, Fe, and Zn). Although there is evidence of the involvement of these metals in AD, further research is needed on their mechanisms of toxicity. To further assess the involvement of heavy and essential metals in AD pathogenesis, we compared cerebrospinal fluid (CSF) AD biomarkers to macro- and microelements measured in CSF and plasma. We tested if macro- and microelements' concentrations (heavy metals (As, Cd, Hg, Ni, Pb, and Tl), essential metals (Na, Mg, K, Ca, Fe, Co, Mn, Cu, Zn, and Mo), essential non-metals (B, P, S, and Se), and other non-essential metals (Al, Ba, Li, and Sr)) are associated with CSF AD biomarkers that reflect pathological changes in the AD brain (amyloid β1-42, total tau, phosphorylated tau isoforms, NFL, S100B, VILIP-1, YKL-40, PAPP-A, and albumin). We used inductively coupled plasma mass spectroscopy (ICP-MS) to determine macro- and microelements in CSF and plasma, and enzyme-linked immunosorbent assays (ELISA) to determine protein biomarkers of AD in CSF. This study included 193 participants (124 with AD, 50 with mild cognitive impairment, and 19 healthy controls). Simple correlation, as well as machine learning algorithms (redescription mining and principal component analysis (PCA)), demonstrated that levels of heavy metals (As, Cd, Hg, Ni, Pb, and Tl), essential metals (Ca, Co, Cu, Fe, Mg, Mn, Mo, Na, K, and Zn), and essential non-metals (P, S, and Se) are positively associated with CSF phosphorylated tau isoforms, VILIP-1, S100B, NFL, and YKL-40 in AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Medical Biology, University of Split School of Medicine, 21000 Split, Croatia
| | - Matej Mihelčić
- Department of Mathematics, University of Zagreb Faculty of Science, 10000 Zagreb, Croatia
| | - Jasna Jurasović
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | | | - Ena Španić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Ankica Sekovanić
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Tatjana Orct
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Klara Zubčić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Nikolina Pleić
- Department of Medical Biology, University of Split School of Medicine, 21000 Split, Croatia
| | | | - Željka Vogrinc
- Laboratory for Neurobiochemistry, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Nela Pivac
- Ruđer Bošković Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia
| | - Andrea Diana
- Laboratory of Neurogenesis and Neuropoiesis, Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Fran Borovečki
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb Medical School, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
43
|
Zou J, Qian J, Liu S, Li R, Zhang X, Yang S, Liu Y, Liu W, Ma S, Shi D. Design, Synthesis, Biological Evaluation and Molecular Dynamics Simulations Study of Genistein‐
O
‐1,3,5‐Triazine Derivatives as Multifunctional Anti‐Alzheimer Agents. ChemistrySelect 2022. [DOI: 10.1002/slct.202203997] [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]
Affiliation(s)
- Jing‐Pei Zou
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
| | - Jing‐Jing Qian
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
| | - Shan‐Ming Liu
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
| | - Rui Li
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
| | - Xiao‐Qing Zhang
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
| | - Shun Yang
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
| | - Yu‐Wei Liu
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology Lianyungang 222005 People's Republic of China
| | - Wei‐Wei Liu
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology Lianyungang 222005 People's Republic of China
| | - Shao‐Jie Ma
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology Lianyungang 222005 People's Republic of China
| | - Da‐Hua Shi
- Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University/Jiangsu Key Laboratory of Marine Bioresources and environment, School of Pharmacy Jiangsu Ocean University Lianyungang 222005 People's Republic of China
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology Lianyungang 222005 People's Republic of China
| |
Collapse
|
44
|
Mahan B, Tacail T, Lewis J, Elliott T, Habekost M, Turner S, Chung R, Moynier F. Exploring the K isotope composition of Göttingen minipig brain regions, and implications for Alzheimer's disease. Metallomics 2022; 14:mfac090. [PMID: 36416864 PMCID: PMC9764214 DOI: 10.1093/mtomcs/mfac090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022]
Abstract
Natural stable metal isotopes have shown utility in differentiation between healthy and diseased brain states (e.g. Alzheimer's disease, AD). While the AD brain accumulates some metals, it purges others, namely K (accompanied by increased serum K, suggesting brain-blood transferal). Here, K isotope compositions of Göttingen minipig brain regions for two AD models at midlife are reported. Results indicate heavy K isotope enrichment where amyloid beta (Aβ) accumulation is observed, and this enrichment correlates with relative K depletion. These results suggest preferential efflux of isotopically light K+ from the brain, a linkage between brain K concentrations and isotope compositions, and linkage to Aβ (previously shown to purge cellular brain K+). Brain K isotope compositions differ from that for serum and brain K is much more abundant than in serum, suggesting that changes in brain K may transfer a measurable K isotope excursion to serum, thereby generating an early AD biomarker.
Collapse
Affiliation(s)
- Brandon Mahan
- IsoTropics Geochemistry Lab, Earth and Environmental Science, James Cook University, Townsville, Queensland 4814, Australia
- Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
- Department of Biomedical Research, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Theo Tacail
- Bristol Isotope Group, School of Earth Sciences, University of Bristol, Bristol BS8 1RJ, UK
- Institute of Geosciences, Johannes Gutenberg University, Mainz 55099, Germany
| | - Jamie Lewis
- Bristol Isotope Group, School of Earth Sciences, University of Bristol, Bristol BS8 1RJ, UK
| | - Tim Elliott
- Bristol Isotope Group, School of Earth Sciences, University of Bristol, Bristol BS8 1RJ, UK
| | - Mette Habekost
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
- Center for Neuroscience, University of Copenhagen Faculty of Health and Medical Sciences, 2200 Copenhagen N, Denmark
| | - Simon Turner
- Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Roger Chung
- Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
- Department of Biomedical Research, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Frédéric Moynier
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, 75238 Paris, France
| |
Collapse
|
45
|
Kumar N, Gupta P, Bansal S. Progress and Development of Carbazole Scaffold Based as Potential Anti-
Alzheimer Agents Using MTDL Approach. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220314144219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Alzheimer’s is a neurodegenerative disease (NDs) found in old age people with associated
most common symptom dementia. MTDLs (Multi-Target Direct Ligand strategy) is based on a combination
of two or more bioactive pharmacophores into a single molecule and this phenomenon has received a
great attention in the new era of modern drug discovery and emerging as a choice to treat this complex
Alzheimer’s disease (AD). In last fifteen years, many research groups designed, and synthesized new
carbazole integrated molecules linked with other bioactive pharmacophores like thiazoles, carvedilol, α-
naphthylaminopropan-2-ol, tacrine, ferulic acid, piperazine, coumarin, chalcones, stilbene, benzyl piperidine,
adamantane, quinoline, phthalocyanines, α-amino phosphonate, thiosemicarbazones, hydrazones,
etc. derivatives using MTDLs approach to confront AD. The present review entails the scientific data on
carbazole hybrids as potential Anti-Alzheimer activities from 2007 to 2021 that have shown potential
anti-Alzheimer activities through multiple target pathways thereby promising hope for new drug development
to confront AD.
Collapse
Affiliation(s)
- Nitin Kumar
- School of Medical and Allied Sciences (SMAS), K.R. Mangalam University, Sohna road, Gurugram, Haryana, India
- Sanskar College of Pharmacy and Research (SCPR), NH-24, Ahead Masuri Canal, Ghaziabad 201302, India
| | - Pankaj Gupta
- School of Medical and Allied Sciences (SMAS), K.R. Mangalam University, Sohna road, Gurugram, Haryana, India
| | - Sahil Bansal
- School of Medical and Allied Sciences (SMAS), K.R. Mangalam University, Sohna road, Gurugram, Haryana, India
| |
Collapse
|
46
|
Abelein A, Ciofi-Baffoni S, Mörman C, Kumar R, Giachetti A, Piccioli M, Biverstål H. Molecular Structure of Cu(II)-Bound Amyloid-β Monomer Implicated in Inhibition of Peptide Self-Assembly in Alzheimer's Disease. JACS AU 2022; 2:2571-2584. [PMID: 36465548 PMCID: PMC9709942 DOI: 10.1021/jacsau.2c00438] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 05/22/2023]
Abstract
Metal ions, such as copper and zinc ions, have been shown to strongly modulate the self-assembly of the amyloid-β (Aβ) peptide into insoluble fibrils, and elevated concentrations of metal ions have been found in amyloid plaques of Alzheimer's patients. Among the physiological transition metal ions, Cu(II) ions play an outstanding role since they can trigger production of neurotoxic reactive oxygen species. In contrast, structural insights into Cu(II) coordination of Aβ have been challenging due to the paramagnetic nature of Cu(II). Here, we employed specifically tailored paramagnetic NMR experiments to determine NMR structures of Cu(II) bound to monomeric Aβ. We found that monomeric Aβ binds Cu(II) in the N-terminus and combined with molecular dynamics simulations, we could identify two prevalent coordination modes of Cu(II). For these, we report here the NMR structures of the Cu(II)-bound Aβ complex, exhibiting heavy backbone RMSD values of 1.9 and 2.1 Å, respectively. Further, applying aggregation kinetics assays, we identified the specific effect of Cu(II) binding on the Aβ nucleation process. Our results show that Cu(II) efficiently retards Aβ fibrillization by predominately reducing the rate of fibril-end elongation at substoichiometric ratios. A detailed kinetic analysis suggests that this specific effect results in enhanced Aβ oligomer generation promoted by Cu(II). These results can quantitatively be understood by Cu(II) interaction with the Aβ monomer, forming an aggregation inert complex. In fact, this mechanism is strikingly similar to other transition metal ions, suggesting a common mechanism of action of retarding Aβ self-assembly, where the metal ion binding to monomeric Aβ is a key determinant.
Collapse
Affiliation(s)
- Axel Abelein
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
| | - Simone Ciofi-Baffoni
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino50019 , Florence, Italy
| | - Cecilia Mörman
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
- Department
of Biochemistry and Biophysics, The Arrhenius Laboratories, Stockholm University, Stockholm106 91, Sweden
| | - Rakesh Kumar
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
| | - Andrea Giachetti
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino50019 , Florence, Italy
| | - Mario Piccioli
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino50019 , Florence, Italy
| | - Henrik Biverstål
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
- Department
of Physical Organic Chemistry, Latvian Institute
of Organic Synthesis, RigaLV-1006, Latvia
| |
Collapse
|
47
|
Summers KL, Roseman G, Schilling KM, Dolgova NV, Pushie MJ, Sokaras D, Kroll T, Harris HH, Millhauser GL, Pickering IJ, George GN. Alzheimer's Drug PBT2 Interacts with the Amyloid β 1-42 Peptide Differently than Other 8-Hydroxyquinoline Chelating Drugs. Inorg Chem 2022; 61:14626-14640. [PMID: 36073854 PMCID: PMC9957665 DOI: 10.1021/acs.inorgchem.2c01694] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although Alzheimer's disease (AD) was first described over a century ago, it remains the leading cause of age-related dementia. Innumerable changes have been linked to the pathology of AD; however, there remains much discord regarding which might be the initial cause of the disease. The "amyloid cascade hypothesis" proposes that the amyloid β (Aβ) peptide is central to disease pathology, which is supported by elevated Aβ levels in the brain before the development of symptoms and correlations of amyloid burden with cognitive impairment. The "metals hypothesis" proposes a role for metal ions such as iron, copper, and zinc in the pathology of AD, which is supported by the accumulation of these metals within amyloid plaques in the brain. Metals have been shown to induce aggregation of Aβ, and metal ion chelators have been shown to reverse this reaction in vitro. 8-Hydroxyquinoline-based chelators showed early promise as anti-Alzheimer's drugs. Both 5-chloro-7-iodo-8-hydroxyquinoline (CQ) and 5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline (PBT2) underwent unsuccessful clinical trials for the treatment of AD. To gain insight into the mechanism of action of 8HQs, we have investigated the potential interaction of CQ, PBT2, and 5,7-dibromo-8-hydroxyquinoline (B2Q) with Cu(II)-bound Aβ(1-42) using X-ray absorption spectroscopy (XAS), high energy resolution fluorescence detected (HERFD) XAS, and electron paramagnetic resonance (EPR). By XAS, we found CQ and B2Q sequestered ∼83% of the Cu(II) from Aβ(1-42), whereas PBT2 sequestered only ∼59% of the Cu(II) from Aβ(1-42), suggesting that CQ and B2Q have a higher relative Cu(II) affinity than PBT2. From our EPR, it became clear that PBT2 sequestered Cu(II) from a heterogeneous mixture of Cu(II)Aβ(1-42) species in solution, leaving a single Cu(II)Aβ(1-42) species. It follows that the Cu(II) site in this Cu(II)Aβ(1-42) species is inaccessible to PBT2 and may be less solvent-exposed than in other Cu(II)Aβ(1-42) species. We found no evidence to suggest that these 8HQs form ternary complexes with Cu(II)Aβ(1-42).
Collapse
Affiliation(s)
- Kelly L. Summers
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham Roseman
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Kevin M. Schilling
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Natalia V. Dolgova
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - M. Jake Pushie
- Department of Surgery, University of Saskatchewan, 103 Hospital Dr, Saskatoon, Saskatchewan S7N 0W8, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Hugh H. Harris
- Department of Chemistry, University of Adelaide, South Australia 5005, Australia
| | - Glenn L. Millhauser
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Ingrid J. Pickering
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham N. George
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
48
|
Liang S, Wu XL, Zong MH, Lou WY. Construction of Zn-heptapeptide bionanozymes with intrinsic hydrolase-like activity for degradation of di(2-ethylhexyl) phthalate. J Colloid Interface Sci 2022; 622:860-870. [PMID: 35561606 DOI: 10.1016/j.jcis.2022.04.122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/28/2022]
Abstract
Nanozyme with intrinsic enzyme-like activity has emerged as favorite artificial catalyst during recent years. However, current nanozymes are mainly limited to inorganic-derived nanomaterials, while biomolecule-sourced nanozyme (bionanozyme) are rarely reported. Herein, inspired by the basic structure of natural hydrolase family, we constructed 3 oligopeptide-based bionanozymes with intrinsic hydrolase-like activity by implementing zinc induced self-assembly of histidine-rich heptapeptides. Under mild condition, divalent zinc (Zn2+) impelled the spontaneous assembly of short peptides (i.e. Ac-IHIHIQI-CONH2, Ac-IHIHIYI-CONH2, and Ac-IHVHLQI-CONH2), forming hydrolase-mimicking bionanozymes with β-sheet secondary conformation and nanofibrous architecture. As expected, the resultant bionanozymes were able to hydrolyze a serious of p-nitrophenyl esters, including not only the simple substrate with short side-chain (p-NPA), but also more complicated ones (p-NPB, p-NPH, p-NPO, and p-NPS). Moreover, the self-assembled Zn-heptapeptide bionanozymes were also proven to be capable of degrading di(2-ethylhexyl) phthalate (DEHP), a typical plasticizer, showing great potential for environmental remediation. Based on this study, we aim to provide theoretical references and exemplify a specific case for directing the construction and application of bionanozyme.
Collapse
Affiliation(s)
- Shan Liang
- Laboratory of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Xiao-Ling Wu
- Laboratory of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Min-Hua Zong
- Laboratory of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Wen-Yong Lou
- Laboratory of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
49
|
Pyun J, McInnes LE, Donnelly PS, Mawal C, Bush AI, Short JL, Nicolazzo JA. Copper bis(thiosemicarbazone) complexes modulate P-glycoprotein expression and function in human brain microvascular endothelial cells. J Neurochem 2022; 162:226-244. [PMID: 35304760 PMCID: PMC9540023 DOI: 10.1111/jnc.15609] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/02/2022]
Abstract
P-glycoprotein (P-gp) is an efflux transporter at the blood-brain barrier (BBB) that hinders brain access of substrate drugs and clears endogenous molecules such as amyloid beta (Aβ) from the brain. As biometals such as copper (Cu) modulate many neuronal signalling pathways linked to P-gp regulation, it was hypothesised that the bis(thiosemicarbazone) (BTSC) Cu-releasing complex, copper II glyoxal bis(4-methyl-3-thiosemicarbazone) (CuII [GTSM]), would enhance P-gp expression and function at the BBB, while copper II diacetyl bis(4-methyl-3-thiosemicarbazone) (CuII [ATSM]), which only releases Cu under hypoxic conditions, would not modulate P-gp expression. Following treatment with 25-250 nM CuII (BTSC)s for 8-48 h, expression of P-gp mRNA and protein in human brain endothelial (hCMEC/D3) cells was assessed by RT-qPCR and Western blot, respectively. P-gp function was assessed by measuring accumulation of the fluorescent P-gp substrate, rhodamine 123 and intracellular Cu levels were quantified by inductively coupled plasma mass spectrometry. Interestingly, CuII (ATSM) significantly enhanced P-gp expression and function 2-fold and 1.3-fold, respectively, whereas CuII (GTSM) reduced P-gp expression 0.5-fold and function by 200%. As both compounds increased intracellular Cu levels, the effect of different BTSC backbones, independent of Cu, on P-gp expression was assessed. However, only the Cu-ATSM complex enhanced P-gp expression and this was mediated partly through activation (1.4-fold) of the extracellular signal-regulated kinase 1 and 2, an outcome that was significantly attenuated in the presence of an inhibitor of the mitogen-activated protein kinase regulatory pathway. Our findings suggest that CuII (ATSM) and CuII (GTSM) have the potential to modulate the expression and function of P-gp at the BBB to impact brain drug delivery and clearance of Aβ.
Collapse
Affiliation(s)
- Jae Pyun
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Lachlan E. McInnes
- Bio21 Molecular Science and Biotechnology InstituteUniversity of MelbourneParkvilleVictoriaAustralia
| | - Paul S. Donnelly
- Bio21 Molecular Science and Biotechnology InstituteUniversity of MelbourneParkvilleVictoriaAustralia
| | - Celeste Mawal
- Oxidation Biology Lab, Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Ashley I. Bush
- Oxidation Biology Lab, Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Jennifer L. Short
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Joseph A. Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| |
Collapse
|
50
|
Lin J, Yang S, Wang Y, Cui Y, Li Q, Chen Y, Ding L. Sensitive detection of levofloxacin and copper (II) based on fluorescence “turn on-off” of biomass carbonized polymer dots. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2022.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|