1
|
Vela Navarro N, De Nadai Mundim G, Cudic M. Implications of Mucin-Type O-Glycosylation in Alzheimer's Disease. Molecules 2025; 30:1895. [PMID: 40363702 PMCID: PMC12073284 DOI: 10.3390/molecules30091895] [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: 03/15/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders linked to aging. Major hallmarks of AD pathogenesis include amyloid-β peptide (Aβ) plaques, which are extracellular deposits originating from the processing of the amyloid precursor protein (APP), and neurofibrillary tangles (NFTs), which are intracellular aggregates of tau protein. Recent evidence indicates that disruptions in metal homeostasis and impaired immune recognition of these aggregates trigger neuroinflammation, ultimately driving disease progression. Therefore, a more comprehensive approach is needed to understand the underlying causes of the disease. Patients with AD present abnormal glycan profiles, and most known AD-related molecules are either modified with glycans or involved in glycan regulation. A deeper understanding of how O-glycosylation influences the balance between amyloid-beta peptide production and clearance, as well as microglia's pro- and anti-inflammatory responses, is crucial for deciphering the early pathogenic events of AD. This review aims to provide a comprehensive summary of the extensive research conducted on the role of mucin-type O-glycosylation in the pathogenesis of AD, discussing its role in disease onset and immune recognition.
Collapse
Affiliation(s)
| | | | - Maré Cudic
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL 33431, USA; (N.V.N.); (G.D.N.M.)
| |
Collapse
|
2
|
Sahoo S, Bandyopadhyay S. Exploring the structure and stability of pentameric amyloid β peptide aggregates in aqueous ammonium-based ionic liquid solutions. Phys Chem Chem Phys 2025; 27:7650-7664. [PMID: 40145365 DOI: 10.1039/d4cp04284k] [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: 03/28/2025]
Abstract
The self-assembly of amyloid beta (Aβ) proteins into fibrils is linked to Alzheimer's disease (AD). Soluble pentamers, particularly those formed in the early stages of Aβ aggregation, are considered highly neurotoxic. This study uses molecular dynamics simulations to explore how trimethylammonium chloride (TMAC), cholinium chloride (ChoC), and tetrabutylammonium chloride (TBAC) ionic liquids (ILs) affect the conformational stability and the association mechanism of Aβ pentameters. These ILs, characterized by varying hydrophilicity/hydrophobicity, exert differential effects on the conformatioanl flexibility of Aβ pentameters. Computational analyses reveal that TBAC induces greater conformational flexibility and multiple energetically favorable states for the Aβ pentamer, potentially driving the pentamerization process along various pathways to form different polymorphic Aβ fibrillar structures. Moreover, analysis of solvent distributions demonstrates that exchange of water by IL ion pairs at the pentamer's exterior surface primarily occurs beyond the first layer of surface-bound water molecules. Particularly, hydrophobic TBA cations show an enhanced propensity to replace weakly interacting water molecules on the surface. Mechanistic insights derived from umbrella sampling simulations further elucidate how ILs modulate the association/dissociation of Aβ monomers within pentameric aggregates. Our findings indicate that the binding of the Aβ peptide becomes less favorable and the binding free energy decreases when transitioning from TMAC to TBAC solutions, as compared to a pure aqueous solution. Finally, energy landscape analysis of Aβ peptide docking to Aβ pentameters reveals multiple low-energy conformations, which are more dispersed in the presence of ChoC and TBAC solutions, potentially hindering Aβ prefibril growth.
Collapse
Affiliation(s)
- Subhadip Sahoo
- Centre for Computational and Data Sciences, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| | - Sanjoy Bandyopadhyay
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur-721302, India.
| |
Collapse
|
3
|
Ongtanasup T, Eawsakul K. Developing Novel Beta-Secretase Inhibitors in a Computer Model as a Possible Treatment for Alzheimer's Disease. Adv Pharmacol Pharm Sci 2025; 2025:5528793. [PMID: 40201042 PMCID: PMC11976051 DOI: 10.1155/adpp/5528793] [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: 08/27/2024] [Accepted: 03/13/2025] [Indexed: 04/10/2025] Open
Abstract
Alzheimer's disease (AD) is a neurological condition that causes neurons and axons in the brain to deteriorate over time and in a specific pattern. The enzyme beta-secretase-1 (BACE-1) plays a crucial role in the onset and progression of AD. In silico approaches, or computer-aided drug design, have become useful tools for reducing the number of therapeutic candidates that need to be evaluated in human clinical trials. Finding chemicals that bind to BACE-1's active site and inhibit its activity is key for preventing AD. A pharmacophore model was developed in this study based on potent BACE-1 inhibitors previously identified, and subsequently employed to screen a commercially available compound database for similar compounds. ZINC35883784 was identified with high binding affinities and hydrogen bonding interactions. Moreover, similar properties to donepezil were found in a compound made by altering the structure of ZINC35883784 called (4R,5R)-2-[1-(2-ethylcyclohexyl)ethyl]-4-hydroxy-5-(4-hydroxybutyl)cyclohexanolate (M4). Compounds were tested for interactions with BACE-1 and favorable properties. Binding scores were confirmed after molecular docking. The assessment of drug-likeness was conducted utilizing Swiss ADME analysis. Molecular dynamics simulations assessed the stability of compound interactions with BACE-1. MMPBSA calculated binding free energy and contribution energy. Results showed that M4 had strong and steady interactions with BACE-1. M4 was also analyzed by predicted NMR and retrosynthesis. However, further experiments are needed to evaluate M4's potential as a BACE-1 inhibitor.
Collapse
Affiliation(s)
- Tassanee Ongtanasup
- Department of Applied Thai Traditional Medicine, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Center of Excellence in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Komgrit Eawsakul
- Department of Applied Thai Traditional Medicine, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
4
|
Berchtold MW, Villalobo A. Ca 2+/calmodulin signaling in organismal aging and cellular senescence: Impact on human diseases. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167583. [PMID: 39579800 DOI: 10.1016/j.bbadis.2024.167583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Molecular mechanisms of aging processes at the level of organisms and cells are in the focus of a large number of research laboratories. This research culminated in recent breakthroughs, which contributed to the better understanding of the natural aging process and aging associated malfunctions leading to age-related diseases. Ca2+ in connection with its master intracellular sensor protein calmodulin (CaM) regulates a plethora of crucial cellular processes orchestrating a wide range of signaling processes. This review focuses on the involvement of Ca2+/CaM in cellular mechanisms, which are associated with normal aging, as well as playing a role in the development of diseases connected with signaling processes during aging. We specifically highlight processes that involve inactivation of proteins, which take part in Ca2+/CaM regulatory systems by oxygen or nitrogen free radical species, during organismal aging and cellular senescence. As examples of organs where aging processes have recently been investigated, we chose to review the literature on molecular aging processes with involvement of Ca2+/CaM in heart and neuronal diseases, as well as in cancer and metabolic diseases, all deeply affected by aging. In addition, this article focuses on cellular senescence, a mechanism that may contribute to aging processes and therefore has been proposed as a target to interfere with the progression of age-associated diseases.
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen Ø, Denmark.
| | - Antonio Villalobo
- Cancer and Human Molecular Genetics Area, Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046 Madrid, Spain.
| |
Collapse
|
5
|
Lim XR, Willemse L, Harraz OF. Amyloid beta Aβ 1-40 activates Piezo1 channels in brain capillary endothelial cells. Biophys J 2024:S0006-3495(24)04106-7. [PMID: 39722451 DOI: 10.1016/j.bpj.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024] Open
Abstract
Amyloid beta (Aβ) peptide accumulation on blood vessels in the brain is a hallmark of neurodegeneration. While Aβ peptides constrict cerebral arteries and arterioles, their impact on capillaries is less understood. Aβ was recently shown to constrict brain capillaries through pericyte contraction, but whether-and if so how-Aβ affects endothelial cells (ECs) remains unknown. ECs represent the predominant vascular cell type in the cerebral circulation, and we recently showed that the mechanosensitive ion channel Piezo1 is functionally expressed in the plasma membrane of ECs. Since Aβ disrupts membrane structures, we hypothesized that Aβ1-40, the predominantly deposited isoform in the cerebral circulation, alters endothelial Piezo1 function. Using patch-clamp electrophysiology and freshly isolated capillary ECs, we assessed the impact of the Aβ1-40 peptide on single-channel Piezo1 activity. We show that Aβ1-40 increased Piezo1 open probability and channel open time. Aβ1-40 effects were absent when Piezo1 was genetically deleted or when a superoxide dismutase/catalase mimetic was used. Further, Aβ1-40 enhanced Piezo1 mechanosensitivity and lowered the pressure of half-maximal Piezo1 activation. Our data collectively suggest that Aβ1-40 facilitates higher Piezo1-mediated cation influx in brain ECs. These novel findings have the potential to unravel the possible involvement of Piezo1 modulation in the pathophysiology of neurodegenerative diseases characterized by Aβ accumulation.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont
| | - Luc Willemse
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont.
| |
Collapse
|
6
|
Zimbone S, Giuffrida ML, Sciacca MFM, Carrotta R, Librizzi F, Milardi D, Grasso G. A VEGF Fragment Encompassing Residues 10-30 Inhibits Aβ1-42 Amyloid Aggregation and Exhibits Neuroprotective Properties Matching the Full-Length Protein. ACS Chem Neurosci 2024; 15:4580-4590. [PMID: 39587417 DOI: 10.1021/acschemneuro.4c00669] [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: 11/27/2024] Open
Abstract
The intricate relationship between brain vascular diseases and neurodegeneration has garnered increased attention in the scientific community. With an aging population, the incidence of these two conditions is likely to increase, making it imperative to understand the underlying common molecular mechanisms and unveiling novel avenues for therapy. Prompted by the observation that Aβ peptide aggregation has been implicated in the development of cerebral amyloid angiopathy (CAA) and that elevated concentrations of vascular endothelial growth factor (VEGF) in the cerebrospinal fluid (CSF) have been correlated with less cognitive decline in Alzheimer's disease (AD), we demonstrate that a small peptide (Pep9) encompassing the 10-30 sequence of VEGF exhibits significant ability to inhibit the aggregation of the Aβ1-42 peptide, as well as the formation of toxic oligomers. AFM studies confirmed this inhibitory capacity, which is also paralleled by a significant reduction of the random coil to a beta-sheet conformational transition. Further studies have shown that Pep9 protects differentiated neuroblastoma SH-SY5Y cells from Aβ toxicity, being even more effective than full-length protein in preventing amyloid-induced neuronal death. The use of a control peptide wherein two histidines are substituted with glycines (H11G and H12G) suggests a close relationship between the peptide amino acid sequence and its antiaggregating/neuroprotective activity. Overall, this study provides insight into the role of VEGF in AD and suggests that specific VEGF fragments could be beneficial in the treatment of this condition.
Collapse
Affiliation(s)
- Stefania Zimbone
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - M Laura Giuffrida
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - Michele F M Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - Rita Carrotta
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Palermo 90146, Italy
| | - Fabio Librizzi
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Palermo 90146, Italy
| | - Danilo Milardi
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - Giulia Grasso
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| |
Collapse
|
7
|
Mohallem R, Schaser AJ, Aryal UK. Molecular Signatures of Neurodegenerative Diseases Identified by Proteomic and Phosphoproteomic Analyses in Aging Mouse Brain. Mol Cell Proteomics 2024; 23:100819. [PMID: 39069073 PMCID: PMC11381985 DOI: 10.1016/j.mcpro.2024.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/05/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024] Open
Abstract
A central hallmark of neurodegenerative diseases is the irreversible accumulation of misfolded proteins in the brain by aberrant phosphorylation. Understanding the mechanisms underlying protein phosphorylation and its role in pathological protein aggregation within the context of aging is crucial for developing therapeutic strategies aimed at preventing or reversing such diseases. Here, we applied multi-protease digestion and quantitative mass spectrometry to compare and characterize dysregulated proteins and phosphosites in the mouse brain proteome using three different age groups: young-adult (3-4 months), middle-age (10 months), and old mice (19-21 months). Proteins associated with senescence, neurodegeneration, inflammation, cell cycle regulation, the p53 hallmark pathway, and cytokine signaling showed significant age-dependent changes in abundances and level of phosphorylation. Several proteins implicated in Alzheimer's disease (AD) and Parkinson's disease (PD) including tau (Mapt), Nefh, and Dpysl2 (also known as Crmp2) were hyperphosphorylated in old mice brain suggesting their susceptibility to the diseases. Cdk5 and Gsk3b, which are known to phosphorylate Dpysl2 at multiple specific sites, had also increased phosphorylation levels in old mice suggesting a potential crosstalk between them to contribute to AD. Hapln2, which promotes α-synuclein aggregation in patients with PD, was one of the proteins with highest abundance in old mice. CD9, which regulates senescence through the PI3K-AKT-mTOR-p53 signaling was upregulated in old mice and its regulation was correlated with the activation of phosphorylated AKT1. Overall, the findings identify a significant association between aging and the dysregulation of proteins involved in various pathways linked to neurodegenerative diseases with potential therapeutic implications.
Collapse
Affiliation(s)
- Rodrigo Mohallem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA
| | - Allison J Schaser
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Uma K Aryal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA; Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
8
|
Wanionok NE, Morel GR, Fernández JM. Osteoporosis and Alzheimer´s disease (or Alzheimer´s disease and Osteoporosis). Ageing Res Rev 2024; 99:102408. [PMID: 38969142 DOI: 10.1016/j.arr.2024.102408] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Alzheimer's disease (AD) and osteoporosis are two diseases that mainly affect elderly people, with increases in the occurrence of cases due to a longer life expectancy. Several epidemiological studies have shown a reciprocal association between both diseases, finding an increase in incidence of osteoporosis in patients with AD, and a higher burden of AD in osteoporotic patients. This epidemiological relationship has motivated the search for molecules, genes, signaling pathways and mechanisms that are related to both pathologies. The mechanisms found in these studies can serve to improve treatments and establish better patient care protocols.
Collapse
Affiliation(s)
- Nahuel E Wanionok
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina
| | - Gustavo R Morel
- Biochemistry Research Institute of La Plata "Professor Doctor Rodolfo R. Brenner" (INIBIOLP), Argentina
| | - Juan M Fernández
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina.
| |
Collapse
|
9
|
Wang Y, Liu S, Spiteri AG, Huynh ALH, Chu C, Masters CL, Goudey B, Pan Y, Jin L. Understanding machine learning applications in dementia research and clinical practice: a review for biomedical scientists and clinicians. Alzheimers Res Ther 2024; 16:175. [PMID: 39085973 PMCID: PMC11293066 DOI: 10.1186/s13195-024-01540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/21/2024] [Indexed: 08/02/2024]
Abstract
Several (inter)national longitudinal dementia observational datasets encompassing demographic information, neuroimaging, biomarkers, neuropsychological evaluations, and muti-omics data, have ushered in a new era of potential for integrating machine learning (ML) into dementia research and clinical practice. ML, with its proficiency in handling multi-modal and high-dimensional data, has emerged as an innovative technique to facilitate early diagnosis, differential diagnosis, and to predict onset and progression of mild cognitive impairment and dementia. In this review, we evaluate current and potential applications of ML, including its history in dementia research, how it compares to traditional statistics, the types of datasets it uses and the general workflow. Moreover, we identify the technical barriers and challenges of ML implementations in clinical practice. Overall, this review provides a comprehensive understanding of ML with non-technical explanations for broader accessibility to biomedical scientists and clinicians.
Collapse
Affiliation(s)
- Yihan Wang
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Shu Liu
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
- The ARC Training Centre in Cognitive Computing for Medical Technologies, The University of Melbourne, Carlton, VIC, 3010, Australia
| | - Alanna G Spiteri
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Andrew Liem Hieu Huynh
- Department of Aged Care, Austin Health, Heidelberg, VIC, 3084, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, 3084, Australia
| | - Chenyin Chu
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Benjamin Goudey
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
- The ARC Training Centre in Cognitive Computing for Medical Technologies, The University of Melbourne, Carlton, VIC, 3010, Australia
| | - Yijun Pan
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.
| | - Liang Jin
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| |
Collapse
|
10
|
Stern S, Crisamore K, Schuck R, Pacanowski M. Evaluation of the landscape of pharmacodynamic biomarkers in Niemann-Pick Disease Type C (NPC). Orphanet J Rare Dis 2024; 19:280. [PMID: 39061081 PMCID: PMC11282650 DOI: 10.1186/s13023-024-03233-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/27/2024] [Indexed: 07/28/2024] Open
Abstract
Niemann-Pick disease type C (NPC) is an autosomal recessive, progressive disorder resulting from variants in NPC1 or NPC2 that leads to the accumulation of cholesterol and other lipids in late endosomes and lysosomes. The clinical manifestations of the disease vary by age of onset, and severity is often characterized by neurological involvement. To date, no disease-modifying therapy has been approved by the United States Food and Drug Administration (FDA) and treatment is typically supportive. The lack of robust biomarkers contributes to challenges associated with disease monitoring and quantifying treatment response. In recent years, advancements in detection methods have facilitated the identification of biomarkers in plasma and cerebral spinal fluid from patients with NPC, namely calbindin D, neurofilament light chain, 24(S)hydroxycholesterol, cholestane-triol, trihydroxycholanic acid glycinate, amyloid-β, total and phosphorylated tau, and N-palmitoyl-O-phosphocholine-serine. These biomarkers have been used to support several clinical trials as pharmacodynamic endpoints. Despite the significant advancements in laboratory techniques, translation of those advancements has lagged, and it remains unclear which biomarkers correlate with disease severity and progression, or which biomarkers could inform treatment response. In this review, we assess the landscape of biomarkers currently proposed to guide disease monitoring or indicate treatment response in patients with NPC.
Collapse
Affiliation(s)
- Sydney Stern
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA.
| | - Karryn Crisamore
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA
| | - Robert Schuck
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA
| | - Michael Pacanowski
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA
| |
Collapse
|
11
|
Rananaware P, Bauri S, Keri R, Mishra M, Brahmkhatri V. Polymeric curcumin nanospheres for lysozyme aggregation inhibition, antibacterial, and wound healing applications. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:46625-46640. [PMID: 37688693 DOI: 10.1007/s11356-023-29160-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/31/2023] [Indexed: 09/11/2023]
Abstract
The present study reports highly stable polymeric nanoparticles comprising curcumin and polyvinylpyrrolidone, and then conjugated with gold nanoparticles, resulting in C-PVP and C-PVP-Au, respectively. The synthesized conjugates C-PVP and C-PVP-Au were investigated for amyloid aggregation inhibition activity, antimicrobial activity, and wound healing applications. The anti-amyloidogenic capacity of nanoconjugates were studied for model protein, hen egg-white lysozyme (HEWL). The ThT binding assay, fibril size measurement, and electron microscopy results revealed that conjugates suppress fibrillogenesis in HEWL. The highest amyloid inhibition activity obtained against C-PVP and C-PVP-Au was 31 μg.mL-1 and 30 μg.mL-1, respectively. The dissociation activity for amyloid aggregation was observed against Q-PVP and Q-PVP-Au at 29 μg.mL-1 and 27 μg.mL-1, respectively. The antibacterial studies show significant efficacy against Escherichia coli (E. coli) in the presence of C-PVP and C-PVP-Au. The substantial antibacterial potential of C-PVP@PVA and C-PVP-Au@PVA membranes shows promising wound healing applications. The PVA membranes with nanoparticles promote the antibacterial activity and wound healing activity in the Drosophila model. C-PVP-Au@PVA membrane healed the wound faster than the C-PVP@PVA, and it can be used for better results in wound healing. Thus, C-PVP-Au and C-PVP have higher bioavailability and stability and can act as multifunctional therapeutic agents for amyloid-related diseases and as wound healing agents. Graphical abstract C-PVP, and C-PVP-Au conjugates for inhibition of HEWL aggregation, antibacterial and wound healing activity.
Collapse
Affiliation(s)
- Pranita Rananaware
- Nanomaterials for Drug Delivery and Therapeutics (NDT-Lab), Centre for Nano and Material Science, Jain University, Jain Global Campus, Bengaluru, 562112, Karnataka, India
| | - Samir Bauri
- Neural Developmental Biology Lab, Department of Life Science, NIT Rourkela, Odisha, Rourkela, 769008, India
| | - Rangappa Keri
- Nanomaterials for Drug Delivery and Therapeutics (NDT-Lab), Centre for Nano and Material Science, Jain University, Jain Global Campus, Bengaluru, 562112, Karnataka, India
| | - Monalisa Mishra
- Neural Developmental Biology Lab, Department of Life Science, NIT Rourkela, Odisha, Rourkela, 769008, India
| | - Varsha Brahmkhatri
- Nanomaterials for Drug Delivery and Therapeutics (NDT-Lab), Centre for Nano and Material Science, Jain University, Jain Global Campus, Bengaluru, 562112, Karnataka, India.
| |
Collapse
|
12
|
Ghosh AK. BACE1 inhibitor drugs for the treatment of Alzheimer's disease: Lessons learned, challenges to overcome, and future prospects †. Glob Health Med 2024; 6:164-168. [PMID: 38947412 PMCID: PMC11197157 DOI: 10.35772/ghm.2024.01033] [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: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Abstract
Alzheimer's disease (AD), first diagnosed over a century ago, remains one of the major healthcare crises around the globe. Currently, there is no cure or effective treatment. The majority of drug development efforts to date have targeted reduction of amyloid-β peptide (Aβ). Drug development through inhibition of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), resulted in promising early clinical studies. However, nearly all small molecule BACE1 inhibitor drugs failed to live up to expectations in later phase clinical trials, due to toxicity and efficacy issues. This commentary aims to provide a brief review of over two decades of BACE1 inhibitor drug development challenges and efforts for treatment of AD and prospects of future BACE1-based drugs.
Collapse
Affiliation(s)
- Arun K. Ghosh
- Departments of Chemistry, Purdue University, West Lafayette, IN, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
13
|
Cheung S, Zhong Y, Wu L, Jia X, He MQ, Ai Y, Jiao Q, Liang Q. Mechanism interpretation of Guhan Yangshengjing for protection against Alzheimer's disease by network pharmacology and molecular docking. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117976. [PMID: 38492794 DOI: 10.1016/j.jep.2024.117976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/20/2024] [Accepted: 02/24/2024] [Indexed: 03/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guhan Yangshengjing (GHYSJ) is an effective prescription for delaying progression of Alzheimer's disease (AD) based on the ancient Chinese medical classics excavated from Mawangdui Han Tomb. Comprising a combination of eleven traditional Chinese herbs, the precise protective mechanism through which GHYSJ acts on AD progression remains unclear and has significant implications for the development of new drugs to treat AD. AIM OF THE STUDY To investigate the mechanism of GHYSJ in the treatment of AD through network pharmacology and validate the results through in vitro experiments. MATERIALS AND METHODS Chemical composition-target-pathway network and protein-protein interaction network were constructed by network pharmacology to predict the potential targets of GHYSJ for the treatment of AD. The interaction relationship between active ingredients and targets was verified by molecular docking and molecular force. Furthermore, the chemical constituents of GHYSJ were analyzed by LC-MS and HPLC, the effects of GHYSJ on animal tissues were analyzed by H&E staining. An Aβ-induced SH-SY5Y cellular model was established to validate the core pathways and targets predicted by network pharmacology and molecular docking. RESULTS The results of the network pharmacology analysis revealed a total of 155 bioactive compounds capable of crossing the blood-brain barrier and interacting with 677 targets, among which 293 targets specifically associated with AD, which mainly participated in and regulated the amyloid aggregation pathway and PI3K/Akt signaling pathway, thereby treating AD. In addition, molecular docking analysis revealed a robust binding affinity between the principal bioactive constituents of GHYSJ and crucial targets implicated in AD. Our findings were further substantiated by in vitro experiments, which demonstrated that Liquiritigenin and Ginsenosides Rh4, crucial constituents of GHYSJ, as well as GHYSJ pharmaceutic serum, exhibited a significant down-regulation of BACE1 expression in Aβ-induced damaged SH-SY5Y cells. This study provides valuable data and theoretical underpinning for the potential therapeutic application of GHYSJ in the treatment of AD and secondary development of GHYSJ prescription. CONCLUSION Through network pharmacology, molecular docking, LC-MS, and cellular experiments, GHYSJ was initially confirmed to delay the progression of AD by regulating the expression of BACE1 in Amyloid aggregation pathway. Our observations provided valuable data and theoretical underpinning for the potential therapeutic application of GHYSJ in the treatment of AD.
Collapse
Affiliation(s)
- Suet Cheung
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, SATCM Key Laboratory of Traditional Chinese Medicine Chemistry, Institute of Traditional Chinese Medicine-X, Chinese Medicine Modernization Research Center, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | | | - Lei Wu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, SATCM Key Laboratory of Traditional Chinese Medicine Chemistry, Institute of Traditional Chinese Medicine-X, Chinese Medicine Modernization Research Center, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xiaomeng Jia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, SATCM Key Laboratory of Traditional Chinese Medicine Chemistry, Institute of Traditional Chinese Medicine-X, Chinese Medicine Modernization Research Center, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Meng-Qi He
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, SATCM Key Laboratory of Traditional Chinese Medicine Chemistry, Institute of Traditional Chinese Medicine-X, Chinese Medicine Modernization Research Center, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, SATCM Key Laboratory of Traditional Chinese Medicine Chemistry, Institute of Traditional Chinese Medicine-X, Chinese Medicine Modernization Research Center, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | | | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, SATCM Key Laboratory of Traditional Chinese Medicine Chemistry, Institute of Traditional Chinese Medicine-X, Chinese Medicine Modernization Research Center, Department of Chemistry, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
14
|
De Meyer S, Schaeverbeke JM, Luckett ES, Reinartz M, Blujdea ER, Cleynen I, Dupont P, Van Laere K, Vanbrabant J, Stoops E, Vanmechelen E, di Molfetta G, Zetterberg H, Ashton NJ, Teunissen CE, Poesen K, Vandenberghe R. Plasma pTau181 and pTau217 predict asymptomatic amyloid accumulation equally well as amyloid PET. Brain Commun 2024; 6:fcae162. [PMID: 39051027 PMCID: PMC11267224 DOI: 10.1093/braincomms/fcae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/25/2024] [Accepted: 05/22/2024] [Indexed: 07/27/2024] Open
Abstract
The dynamic phase of preclinical Alzheimer's disease, as characterized by accumulating cortical amyloid-β, is a window of opportunity for amyloid-β-lowering therapies to have greater efficacy. Biomarkers that accurately predict amyloid-β accumulation may be of critical importance for participant inclusion in secondary prevention trials and thus enhance development of early Alzheimer's disease therapies. We compared the abilities of baseline plasma pTau181, pTau217 and amyloid-β PET load to predict future amyloid-β accumulation in asymptomatic elderly. In this longitudinal cohort study, baseline plasma pTau181 and pTau217 were quantified using single molecule array assays in cognitively unimpaired elderly selected from the community-recruited F-PACK cohort based on the availability of baseline plasma samples and longitudinal amyloid-β PET data (median time interval = 5 years, range 2-10 years). The predictive abilities of pTau181, pTau217 and PET-based amyloid-β measures for PET-based amyloid-β accumulation were investigated using receiver operating characteristic analyses, correlations and stepwise regression analyses. We included 75 F-PACK subjects (mean age = 70 years, 48% female), of which 16 were classified as amyloid-β accumulators [median (interquartile range) Centiloid rate of change = 3.42 (1.60) Centiloids/year). Plasma pTau181 [area under the curve (95% confidence interval) = 0.72 (0.59-0.86)] distinguished amyloid-β accumulators from non-accumulators with similar accuracy as pTau217 [area under the curve (95% confidence interval) = 0.75 (0.62-0.88) and amyloid-β PET [area under the curve (95% confidence interval) = 0.72 (0.56-0.87)]. Plasma pTau181 and pTau217 strongly correlated with each other (r = 0.93, Pfalse discovery rate < 0.001) and, together with amyloid-β PET, similarly correlated with amyloid-β rate of change (r pTau181 = 0.33, r pTau217 = 0.36, r amyloid-β PET = 0.35, all Pfalse discovery rate ≤ 0.01). Addition of plasma pTau181, plasma pTau217 or amyloid-β PET to a linear demographic model including age, sex and APOE-ε4 carriership similarly improved the prediction of amyloid-β accumulation (ΔAkaike information criterion ≤ 4.1). In a multimodal biomarker model including all three biomarkers, each biomarker lost their individual predictive ability. These findings indicate that plasma pTau181, plasma pTau217 and amyloid-β PET convey overlapping information and therefore predict the dynamic phase of asymptomatic amyloid-β accumulation with comparable performances. In clinical trial recruitment, confirmatory PET scans following blood-based prescreening might thus not provide additional value for detecting participants in these early disease stages who are destined to accumulate cortical amyloid-β. Given the moderate performances, future studies should investigate whether integrating plasma pTau species with other factors can improve performance and thus enhance clinical and research utility.
Collapse
Affiliation(s)
- Steffi De Meyer
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Jolien M Schaeverbeke
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Emma S Luckett
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Laboratory for Complex Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Mariska Reinartz
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Elena R Blujdea
- Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HV Amsterdam, The Netherlands
| | - Isabelle Cleynen
- Laboratory for Complex Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Patrick Dupont
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
- Division of Nuclear Medicine, UZ Leuven, 3000 Leuven, Belgium
| | | | | | | | - Guglielmo di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
- UK Dementia Research Institute at UCL, WC1N 3BG London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HV Amsterdam, The Netherlands
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Laboratory Medicine Department, UZ Leuven, 3000 Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Neurology Department, UZ Leuven, 3000 Leuven, Belgium
| |
Collapse
|
15
|
Varenyk Y, Theodorakis PE, Pham DQH, Li MS, Krupa P. Exploring Structural Insights of Aβ42 and α-Synuclein Monomers and Heterodimer: A Comparative Study Using Implicit and Explicit Solvent Simulations. J Phys Chem B 2024; 128:4655-4669. [PMID: 38700150 PMCID: PMC11103699 DOI: 10.1021/acs.jpcb.4c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024]
Abstract
Protein misfolding, aggregation, and fibril formation play a central role in the development of severe neurological disorders, including Alzheimer's and Parkinson's diseases. The structural stability of mature fibrils in these diseases is of great importance, as organisms struggle to effectively eliminate amyloid plaques. To address this issue, it is crucial to investigate the early stages of fibril formation when monomers aggregate into small, toxic, and soluble oligomers. However, these structures are inherently disordered, making them challenging to study through experimental approaches. Recently, it has been shown experimentally that amyloid-β 42 (Aβ42) and α-synuclein (α-Syn) can coassemble. This has motivated us to investigate the interaction between their monomers as a first step toward exploring the possibility of forming heterodimeric complexes. In particular, our study involves the utilization of various Amber and CHARMM force-fields, employing both implicit and explicit solvent models in replica exchange and conventional simulation modes. This comprehensive approach allowed us to assess the strengths and weaknesses of these solvent models and force fields in comparison to experimental and theoretical findings, ensuring the highest level of robustness. Our investigations revealed that Aβ42 and α-Syn monomers can indeed form stable heterodimers, and the resulting heterodimeric model exhibits stronger interactions compared to the Aβ42 dimer. The binding of α-Syn to Aβ42 reduces the propensity of Aβ42 to adopt fibril-prone conformations and induces significant changes in its conformational properties. Notably, in AMBER-FB15 and CHARMM36m force fields with the use of explicit solvent, the presence of Aβ42 significantly increases the β-content of α-Syn, consistent with the experiments showing that Aβ42 triggers α-Syn aggregation. Our analysis clearly shows that although the use of implicit solvent resulted in too large compactness of monomeric α-Syn, structural properties of monomeric Aβ42 and the heterodimer were preserved in explicit-solvent simulations. We anticipate that our study sheds light on the interaction between α-Syn and Aβ42 proteins, thus providing the atom-level model required to assess the initial stage of aggregation mechanisms related to Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Yuliia Varenyk
- Institute
of Physics Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
- Department
of Theoretical Chemistry, University of
Vienna, Vienna 1090, Austria
| | | | - Dinh Q. H. Pham
- Institute
of Physics Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Mai Suan Li
- Institute
of Physics Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Paweł Krupa
- Institute
of Physics Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| |
Collapse
|
16
|
Shrestha HK, Lee D, Wu Z, Wang Z, Fu Y, Wang X, Serrano GE, Beach TG, Peng J. Profiling Protein-Protein Interactions in the Human Brain by Refined Cofractionation Mass Spectrometry. J Proteome Res 2024; 23:1221-1231. [PMID: 38507900 PMCID: PMC11065482 DOI: 10.1021/acs.jproteome.3c00685] [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] [Indexed: 03/22/2024]
Abstract
Proteins usually execute their biological functions through interactions with other proteins and by forming macromolecular complexes, but global profiling of protein complexes directly from human tissue samples has been limited. In this study, we utilized cofractionation mass spectrometry (CF-MS) to map protein complexes within the postmortem human brain with experimental replicates. First, we used concatenated anion and cation Ion Exchange Chromatography (IEX) to separate native protein complexes in 192 fractions and then proceeded with Data-Independent Acquisition (DIA) mass spectrometry to analyze the proteins in each fraction, quantifying a total of 4,804 proteins with 3,260 overlapping in both replicates. We improved the DIA's quantitative accuracy by implementing a constant amount of bovine serum albumin (BSA) in each fraction as an internal standard. Next, advanced computational pipelines, which integrate both a database-based complex analysis and an unbiased protein-protein interaction (PPI) search, were applied to identify protein complexes and construct protein-protein interaction networks in the human brain. Our study led to the identification of 486 protein complexes and 10054 binary protein-protein interactions, which represents the first global profiling of human brain PPIs using CF-MS. Overall, this study offers a resource and tool for a wide range of human brain research, including the identification of disease-specific protein complexes in the future.
Collapse
Affiliation(s)
- Him K. Shrestha
- Departments of Structural Biology and Developmental Neurobiology
| | - DongGeun Lee
- Departments of Structural Biology and Developmental Neurobiology
| | - Zhiping Wu
- Departments of Structural Biology and Developmental Neurobiology
| | - Zhen Wang
- Departments of Structural Biology and Developmental Neurobiology
| | - Yingxue Fu
- Departments of Structural Biology and Developmental Neurobiology
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, Tennessee, 38105, USA
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, Tennessee, 38105, USA
| | | | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology
| |
Collapse
|
17
|
Dey P, Biswas P. Effect of caffeine on the aggregation of amyloid-β-A 3D RISM study. J Chem Phys 2024; 160:125101. [PMID: 38516974 DOI: 10.1063/5.0202636] [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: 02/06/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
Alzheimer's disease is a detrimental neurological disorder caused by the formation of amyloid fibrils due to the aggregation of amyloid-β peptide. The primary therapeutic approaches for treating Alzheimer's disease are targeted to prevent this amyloid fibril formation using potential inhibitor molecules. The discovery of such inhibitor molecules poses a formidable challenge to the design of anti-amyloid drugs. This study investigates the effect of caffeine on dimer formation of the full-length amyloid-β using a combined approach of all-atom, explicit water molecular dynamics simulations and the three-dimensional reference interaction site model theory. The change in the hydration free energy of amyloid-β dimer, with and without the inhibitor molecules, is calculated with respect to the monomeric amyloid-β, where the hydration free energy is decomposed into energetic and entropic components, respectively. Dimerization is accompanied by a positive change in the partial molar volume. Dimer formation is spontaneous, which implies a decrease in the hydration free energy. However, a reverse trend is observed for the dimer with inhibitor molecules. It is observed that the negatively charged residues primarily contribute for the formation of the amyloid-β dimer. A residue-wise decomposition reveals that hydration/dehydration of the side-chain atoms of the charged amino acid residues primarily contribute to dimerization.
Collapse
Affiliation(s)
- Priya Dey
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Parbati Biswas
- Department of Chemistry, University of Delhi, Delhi 110007, India
| |
Collapse
|
18
|
Umoh IO, dos Reis HJ, de Oliveira ACP. Molecular Mechanisms Linking Osteoarthritis and Alzheimer's Disease: Shared Pathways, Mechanisms and Breakthrough Prospects. Int J Mol Sci 2024; 25:3044. [PMID: 38474288 PMCID: PMC10931612 DOI: 10.3390/ijms25053044] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease mostly affecting the elderly population. It is characterized by cognitive decline that occurs due to impaired neurotransmission and neuronal death. Even though deposition of amyloid beta (Aβ) peptides and aggregation of hyperphosphorylated TAU have been established as major pathological hallmarks of the disease, other factors such as the interaction of genetic and environmental factors are believed to contribute to the development and progression of AD. In general, patients initially present mild forgetfulness and difficulty in forming new memories. As it progresses, there are significant impairments in problem solving, social interaction, speech and overall cognitive function of the affected individual. Osteoarthritis (OA) is the most recurrent form of arthritis and widely acknowledged as a whole-joint disease, distinguished by progressive degeneration and erosion of joint cartilage accompanying synovitis and subchondral bone changes that can prompt peripheral inflammatory responses. Also predominantly affecting the elderly, OA frequently embroils weight-bearing joints such as the knees, spine and hips leading to pains, stiffness and diminished joint mobility, which in turn significantly impacts the patient's standard of life. Both infirmities can co-occur in older adults as a result of independent factors, as multiple health conditions are common in old age. Additionally, risk factors such as genetics, lifestyle changes, age and chronic inflammation may contribute to both conditions in some individuals. Besides localized peripheral low-grade inflammation, it is notable that low-grade systemic inflammation prompted by OA can play a role in AD pathogenesis. Studies have explored relationships between systemic inflammatory-associated diseases like obesity, hypertension, dyslipidemia, diabetes mellitus and AD. Given that AD is the most common form of dementia and shares similar risk factors with OA-both being age-related and low-grade inflammatory-associated diseases, OA may indeed serve as a risk factor for AD. This work aims to review literature on molecular mechanisms linking OA and AD pathologies, and explore potential connections between these conditions alongside future prospects and innovative treatments.
Collapse
Affiliation(s)
| | - Helton Jose dos Reis
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil;
| | - Antonio Carlos Pinheiro de Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil;
| |
Collapse
|
19
|
Dahdah A, de Silva NH, Maniam S, Blanch EW. Characterizing fibril morphological changes by spirooxindoles for neurodegenerative disease application. Analyst 2024; 149:1229-1237. [PMID: 38224234 DOI: 10.1039/d3an01773g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Fibrillation of proteins and polypeptides, which leads to the deposition of plaques in cells and tissues has been widely associated with many neuropathological diseases. Inhibition of protein misfolding and aggregation is crucial for the prevention and treatment of these conditions. The growing interest in identifying inhibitor molecules to prevent the formation of fibrils in vivo has led to the results highlighted in this study. Due to their hydrophobic structure and potential to readily cross the blood brain barrier, a library of spirooxindole compounds were synthesized with those labelled Hd-63, Hd-66 and Hd-74 proving to be the most potent against fibril formation. Our spectroscopic analysis provides detailed insight, that the introduction of these spirooxindole compounds leads to morphological changes in the mechanism of fibril formation which prevent the formation of highly ordered fibrils, instead results in the formation of disordered aggregates which are not fibrillar in nature.
Collapse
Affiliation(s)
- Anthony Dahdah
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia.
| | - Nilamuni H de Silva
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia.
| | - Subashani Maniam
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia.
| | - Ewan W Blanch
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia.
| |
Collapse
|
20
|
Yokoyama K, Barbour E, Hirschkind R, Martinez Hernandez B, Hausrath K, Lam T. Protein Corona Formation and Aggregation of Amyloid β 1-40-Coated Gold Nanocolloids. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:1728-1746. [PMID: 38194428 DOI: 10.1021/acs.langmuir.3c02923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Amyloid fibrillogenesis is a pathogenic protein aggregation process that occurs through a highly ordered process of protein-protein interactions. To better understand the protein-protein interactions involved in amyloid fibril formation, we formed nanogold colloid aggregates by stepwise additions of ∼2 nmol of amyloid β 1-40 peptide (Aβ1-40) at pH ∼3.7 and ∼25 °C. The processes of protein corona formation and building of gold colloid [diameters (d) of 20 and 80 nm] aggregates were confirmed by a red-shift of the surface plasmon resonance (SPR) band, λpeak, as the number of Aβ1-40 peptides [N(Aβ1-40)] increased. The normalized red-shift of λpeak, Δλ, was correlated with the degree of protein aggregation, and this process was approximated as the adsorption isotherm explained by the Langmuir-Freundlich model. As the coverage fraction (θ) was analyzed as a function of ϕ, which is the N(Aβ1-40) per total surface area of nanogold colloids available for adsorption, the parameters for explaining the Langmuir-Freundlich model were in good agreement for both 20 and 80 nm gold, indicating that ϕ could define the stage of the aggregation process. Surface-enhanced Raman scattering (SERS) imaging was conducted at designated values of ϕ and suggested that a protein-gold surface interaction during the initial adsorption stage may be dependent on the nanosize. The 20 nm gold case seems to prefer a relatively smaller contacting section, such as a -C-N or C═C bond, but a plane of the benzene ring may play a significant role for 80 nm gold. Regardless of the size of the particles, the β-sheet and random coil conformations were considered to be used to form gold colloid aggregates. The methodology developed in this study allows for new insights into protein-protein interactions at distinct stages of aggregation.
Collapse
Affiliation(s)
- Kazushige Yokoyama
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Eli Barbour
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Rachel Hirschkind
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Bryan Martinez Hernandez
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Kaylee Hausrath
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Theresa Lam
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| |
Collapse
|
21
|
Seol SI, Davaanyam D, Oh SA, Lee EH, Han PL, Kim SW, Lee JK. Age-Dependent and Aβ-Induced Dynamic Changes in the Subcellular Localization of HMGB1 in Neurons and Microglia in the Brains of an Animal Model of Alzheimer's Disease. Cells 2024; 13:189. [PMID: 38247880 PMCID: PMC10814163 DOI: 10.3390/cells13020189] [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: 09/27/2023] [Revised: 12/12/2023] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
HMGB1 is a prototypical danger-associated molecular pattern (DAMP) molecule that co-localizes with amyloid beta (Aβ) in the brains of patients with Alzheimer's disease. HMGB1 levels are significantly higher in the cerebrospinal fluid of patients. However, the cellular and subcellular distribution of HMGB1 in relation to the pathology of Alzheimer's disease has not yet been studied in detail. Here, we investigated whether HMGB1 protein levels in brain tissue homogenates (frontal cortex and striatum) and sera from Tg-APP/PS1 mice, along with its cellular and subcellular localization in those regions, differed. Total HMGB1 levels were increased in the frontal cortices of aged wildtype (7.5 M) mice compared to young (3.5 M) mice, whereas total HMGB1 levels in the frontal cortices of Tg-APP/PS1 mice (7.5 M) were significantly lower than those in age-matched wildtype mice. In contrast, total serum HMGB1 levels were enhanced in aged wildtype (7.5 M) mice and Tg-APP/PS1 mice (7.5 M). Further analysis indicated that nuclear HMGB1 levels in the frontal cortices of Tg-APP/PS1 mice were significantly reduced compared to those in age-matched wildtype controls, and cytosolic HMGB1 levels were also significantly decreased. Triple-fluorescence immunohistochemical analysis indicated that HMGB1 appeared as a ring shape in the cytoplasm of most neurons and microglia in the frontal cortices of 9.5 M Tg-APP/PS1 mice, indicating that nuclear HMGB1 is reduced by aging and in Tg-APP/PS1 mice. Consistent with these observations, Aβ treatment of both primary cortical neuron and primary microglial cultures increased HMGB1 secretion in the media, in an Aβ-dose-dependent manner. Our results indicate that nuclear HMGB1 might be translocated from the nucleus to the cytoplasm in both neurons and microglia in the brains of Tg-APP/PS1 mice, and that it may subsequently be secreted extracellularly.
Collapse
Affiliation(s)
- Song-I Seol
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Republic of Korea; (S.-I.S.); (D.D.); (S.-A.O.)
| | - Dashdulam Davaanyam
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Republic of Korea; (S.-I.S.); (D.D.); (S.-A.O.)
| | - Sang-A Oh
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Republic of Korea; (S.-I.S.); (D.D.); (S.-A.O.)
| | - Eun-Hwa Lee
- Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul 03760, Republic of Korea; (E.-H.L.); (P.-L.H.)
| | - Pyung-Lim Han
- Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul 03760, Republic of Korea; (E.-H.L.); (P.-L.H.)
- Department of Chemistry and Nano Science, College of Natural Science, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seung-Woo Kim
- Department of Biomedical Sciences, Inha University School of Medicine, Inchon 22212, Republic of Korea
| | - Ja-Kyeong Lee
- Department of Anatomy, Inha University School of Medicine, Incheon 22212, Republic of Korea; (S.-I.S.); (D.D.); (S.-A.O.)
| |
Collapse
|
22
|
Wolf EJ, Miller MW, Hawn SE, Zhao X, Wallander SE, McCormick B, Govan C, Rasmusson A, Stone A, Schichman SA, Logue MW. Longitudinal study of traumatic-stress related cellular and cognitive aging. Brain Behav Immun 2024; 115:494-504. [PMID: 37967663 PMCID: PMC10843744 DOI: 10.1016/j.bbi.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/18/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023] Open
Abstract
Traumatic stress is associated with both accelerated epigenetic age and increased risk for dementia. Accelerated epigenetic age might link symptoms of traumatic stress to dementia-associated biomarkers, such as amyloid-beta (Aβ) proteins, neurofilament light (NFL), and inflammatory molecules. We tested this hypothesis using longitudinal data obtained from 214 trauma-exposed military veterans (85 % male, mean age at baseline: 53 years, 75 % White) who were assessed twice over the course of an average of 5.6 years. Cross-lagged panel mediation models evaluated measures of lifetime posttraumatic stress disorder and internalizing and externalizing comorbidity (assessed at Time 1; T1) in association with T1 epigenetic age (per the GrimAge algorithm) and T1 plasma markers of neuropathology along with bidirectional temporal paths between T1 and T2 epigenetic age and the plasma markers. Results revealed that a measure of externalizing comorbidity was associated with accelerated epigenetic age (β = 0.30, p <.01), which in turn, was associated with subsequent increases in Aβ-40 (β = 0.20, p <.001), Aβ-42 (β = 0.18, p <.001), and interleukin-6 (β = 0.18, p <.01). T1 advanced epigenetic age and the T1 neuropathology biomarkers NFL and glial fibrillary acidic protein predicted worse performance on T2 neurocognitive tasks assessing working memory, executive/attentional control, and/or verbal memory (ps = 0.03 to 0.009). Results suggest that advanced GrimAge is predictive of subsequent increases in neuropathology and inflammatory biomarkers as well as worse cognitive function, highlighting the clinical significance of this biomarker with respect to cognitive aging and brain health over time. The finding that advanced GrimAge mediated the association between psychiatric comorbidity and future neuropathology is important for understanding potential pathways to neurodegeneration and early identification of those at greatest risk.
Collapse
Affiliation(s)
- Erika J Wolf
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA.
| | - Mark W Miller
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Sage E Hawn
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Old Dominion University, Department of Psychology, Norfolk, VA, USA
| | - Xiang Zhao
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Sara E Wallander
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Beth McCormick
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Christine Govan
- MAVERIC Central Biorepository, VA Boston Healthcare System, Boston, MA, USA
| | - Ann Rasmusson
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA
| | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Steven A Schichman
- Pathology and Laboratory Medicine Service, Central Arkansas Veterans Healthcare System, USA; Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mark W Logue
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA; Boston University Chobanian & Avedisian School of Medicine, Department of Psychiatry, Boston, MA, USA; Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA; Boston University School of Medicine, Department of Medicine, Biomedical Genetics, Boston, MA, USA
| |
Collapse
|
23
|
Xiong R, Li B, Yu H, Fan T, Yu H, Yang Y, Wang JZ, Pi G, Yang X. Urolithin A Inhibits Anterior Basolateral Amygdala to Ventral Hippocampal CA1 Circuit to Ameliorate Amyloid-β-Impaired Social Ability. J Alzheimers Dis 2024; 99:1303-1316. [PMID: 38759018 DOI: 10.3233/jad-240298] [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: 05/19/2024]
Abstract
BACKGROUND Anxiety and social withdrawal are highly prevalent among patients with Alzheimer's disease (AD). However, the neural circuit mechanisms underlying these symptoms remain elusive, and there is a need for effective prevention strategies. OBJECTIVE This study aims to elucidate the neural circuitry mechanisms underlying social anxiety in AD. METHODS We utilized 5xFAD mice and conducted a series of experiments including optogenetic manipulation, Tandem Mass Tag-labeled proteome analysis, behavioral assessments, and immunofluorescence staining. RESULTS In 5xFAD mice, we observed significant amyloid-β (Aβ) accumulation in the anterior part of basolateral amygdala (aBLA). Behaviorally, 6-month-old 5xFAD mice displayed excessive social avoidance during social interaction. Concurrently, the pathway from aBLA to ventral hippocampal CA1 (vCA1) was significantly activated and exhibited a disorganized firing patterns during social interaction. By optogenetically inhibiting the aBLA-vCA1 pathway, we effectively improved the social ability of 5xFAD mice. In the presence of Aβ accumulation, we identified distinct changes in the protein network within the aBLA. Following one month of administration of Urolithin A (UA), we observed significant restoration of the abnormal protein network within the aBLA. UA treatment also attenuated the disorganized firings of the aBLA-vCA1 pathway, leading to an improvement in social ability. CONCLUSIONS The aBLA-vCA1 circuit is a vulnerable pathway in response to Aβ accumulation during the progression of AD and plays a crucial role in Aβ-induced social anxiety. Targeting the aBLA-vCA1 circuit and UA administration are both effective strategies for improving the Aβ-impaired social ability.
Collapse
Affiliation(s)
- Rui Xiong
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binrui Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haitao Yu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Fundamental Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Tianceng Fan
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guilin Pi
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
24
|
Sharma H, Dar TA, Wijayasinghe YS, Sahoo D, Poddar NK. Nano-Osmolyte Conjugation: Tailoring the Osmolyte-Protein Interactions at the Nanoscale. ACS OMEGA 2023; 8:47367-47379. [PMID: 38144115 PMCID: PMC10733987 DOI: 10.1021/acsomega.3c07248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 12/26/2023]
Abstract
Osmolytes are small organic compounds accumulated at higher concentrations in the cell under various stress conditions like high temperature, high salt, high pressure, etc. Osmolytes mainly include four major classes of compounds including sugars, polyols, methylamines, and amino acids and their derivatives. In addition to their ability to maintain protein stability and folding, these osmolytes, also termed as chemical chaperones, can prevent protein misfolding and aggregation. Although being efficient protein folders and stabilizers, these osmolytes exhibit certain unavoidable limitations such as nearly molar concentrations of osmolytes being required for their effect, which is quite difficult to achieve inside a cell or in the extracellular matrix due to nonspecificity and limited permeability of the blood-brain barrier system and reduced bioavailability. These limitations can be overcome to a certain extent by using smart delivery platforms for the targeted delivery of osmolytes to the site of action. In this context, osmolyte-functionalized nanoparticles, termed nano-osmolytes, enhance the protein stabilization and chaperone efficiency of osmolytes up to 105 times in certain cases. For example, sugars, polyols, and amino acid functionalized based nano-osmolytes have shown tremendous potential in preventing protein aggregation. The enhanced potential of nano-osmolytes can be attributed to their high specificity at low concentrations, high tunability, amphiphilicity, multivalent complex formation, and efficient drug delivery system. Keeping in view the promising potential of nano-osmolytes conjugation in tailoring the osmolyte-protein interactions, as compared to their molecular forms, the present review summarizes the recent advancements of the nano-osmolytes that enhance the protein stability/folding efficiency and ability to act as artificial chaperones with increased potential to prevent protein misfolding disorders. Some of the potential nano-osmolyte aggregation inhibitors have been highlighted for large-scale screening with future applications in aggregation disorders. The synthesis of nano-osmolytes by numerous approaches and future perspectives are also highlighted.
Collapse
Affiliation(s)
- Hemlata Sharma
- Department
of Biosciences, Manipal University Jaipur, Jaipur-Ajmer Express Highway, Dehmi
Kalan, Near GVK Toll Plaza, Jaipur, Rajasthan 303007, India
| | - Tanveer Ali Dar
- Department
of Clinical Biochemistry, University of
Kashmir, Srinagar 190006, Jammu and Kashmir India
| | | | - Dibakar Sahoo
- School
of Physics, Sambalpur University, Jyoti Vihar, Burla 768019, Odisha, India
| | - Nitesh Kumar Poddar
- Department
of Biosciences, Manipal University Jaipur, Jaipur-Ajmer Express Highway, Dehmi
Kalan, Near GVK Toll Plaza, Jaipur, Rajasthan 303007, India
| |
Collapse
|
25
|
Piccialli I, Greco F, Roviello G, Sisalli MJ, Tedeschi V, di Mola A, Borbone N, Oliviero G, De Feo V, Secondo A, Massa A, Pannaccione A. The 3-(3-oxoisoindolin-1-yl)pentane-2,4-dione (ISOAC1) as a new molecule able to inhibit Amyloid β aggregation and neurotoxicity. Biomed Pharmacother 2023; 168:115745. [PMID: 37871561 DOI: 10.1016/j.biopha.2023.115745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
Amyloid β 1-42 (Aβ1-42) protein aggregation is considered one of the main triggers of Alzheimer's disease (AD). In this study, we examined the in vitro anti-amyloidogenic activity of the isoindolinone derivative 3-(3-oxoisoindolin-1-yl)pentane-2,4-dione (ISOAC1) and its neuroprotective potential against the Aβ1-42 toxicity. By performing the Thioflavin T fluorescence assay, Western blotting analyses, and Circular Dichroism experiments, we found that ISOAC1 was able to reduce the Aβ1-42 aggregation and conformational transition towards β-sheet structures. Interestingly, in silico studies revealed that ISOAC1 was able to bind to both the monomer and a pentameric protofibril of Aβ1-42, establishing a hydrophobic interaction with the PHE19 residue of the Aβ1-42 KLVFF motif. In vitro analyses on primary cortical neurons showed that ISOAC1 counteracted the increase of intracellular Ca2+ levels and decreased the Aβ1-42-induced toxicity, in terms of mitochondrial activity reduction and increase of reactive oxygen species production. In addition, confocal microscopy analyses showed that ISOAC1 was able to reduce the Aβ1-42 intraneuronal accumulation. Collectively, our results clearly show that ISOAC1 exerts a neuroprotective effect by reducing the Aβ1-42 aggregation and toxicity, hence emerging as a promising compound for the development of new Aβ-targeting therapeutic strategies for AD treatment.
Collapse
Affiliation(s)
- Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Francesca Greco
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Giovanni Roviello
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Naples, Italy
| | - Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Antonia di Mola
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano, SA, Italy
| | - Nicola Borbone
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Giorgia Oliviero
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples, Naples, Italy
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Antonio Massa
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano, SA, Italy.
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy.
| |
Collapse
|
26
|
Han YL, Yin HH, Xiao C, Bernards MT, He Y, Guan YX. Understanding the Molecular Mechanisms of Polyphenol Inhibition of Amyloid β Aggregation. ACS Chem Neurosci 2023; 14:4051-4061. [PMID: 37890131 DOI: 10.1021/acschemneuro.3c00586] [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: 10/29/2023] Open
Abstract
Alzheimer's disease (AD) is highly associated with self-aggregation of amyloid β (Aβ) proteins into fibrils. Inhibition of Aβ aggregation by polyphenols is one of the major therapeutic strategies for AD. Among them, four polyphenols (brazilin, resveratrol, hematoxylin, and rosmarinic acid) have been reported to be effective at inhibiting Aβ aggregation, but the inhibition mechanisms are still unclear. In this work, these four polyphenols were selected to explore their interactions with the Aβ17-42 pentamer by molecular dynamics simulation. All four polyphenols can bind to the pentamer tightly but prefer different binding sites. Conversion of the β-sheet to the random coil, fewer interchain hydrogen bonds, and weaker salt bridges were observed after binding. Interestingly, different Aβ17-42 pentamer destabilizing mechanisms for resveratrol and hematoxylin were found. Resveratrol inserts into the hydrophobic core of the pentamer by forming hydrogen bonds with Asp23 and Lys28, while hematoxylin prefers to bind beside chain A of the pentamer, which leads to β-sheet offset and dissociation of the β1 sheet of chain E. This work reveals the interactions between the Aβ17-42 pentamer and four polyphenols and discusses the relationship between inhibitor structures and their inhibition mechanisms, which also provides useful guidance for screening effective Aβ aggregation inhibitors and drug design against AD.
Collapse
Affiliation(s)
- Yin-Lei Han
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Huan-Huan Yin
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Chao Xiao
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Matthew T Bernards
- Department of Chemical and Biological Engineering, University of Idaho, Moscow 83844, Idaho, United States
| | - Yi He
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Chemical Engineering, University of Washington, Seattle 98195, Washington, United States
| | - Yi-Xin Guan
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
27
|
Kuang S, Zhu B, Zhang J, Yang F, Wu B, Ding W, Yang L, Shen S, Liang SH, Mondal P, Kumar M, Tanzi RE, Zhang C, Chao H, Ran C. A Photolabile Curcumin-Diazirine Analogue Enables Phototherapy with Physically and Molecularly Produced Light for Alzheimer's Disease Treatment. Angew Chem Int Ed Engl 2023; 62:e202312519. [PMID: 37721455 PMCID: PMC10615883 DOI: 10.1002/anie.202312519] [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: 08/25/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/19/2023]
Abstract
The development of Alzheimer's disease (AD) drugs has recently witnessed substantial achievement. To further enhance the pool of drug candidates, it is crucial to explore non-traditional therapeutic avenues. In this study, we present the use of a photolabile curcumin-diazirine analogue, CRANAD-147, to induce changes in properties, structures (sequences), and neurotoxicity of amyloid beta (Aβ) species both in cells and in vivo. This manipulation was achieved through irradiation with LED light or molecularly generated light, dubbed as "molecular light", emitted by the chemiluminescence probe ADLumin-4. Next, aided by molecular chemiluminescence imaging, we demonstrated that the combination of CRANAD-147/LED or CRANAD-147/ADLumin-4 (molecular light) could effectively slow down the accumulation of Aβs in transgenic 5xFAD mice in vivo. Leveraging the remarkable tissue penetration capacity of molecular light, phototherapy employing the synergistic effect of a photolabile Aβ ligand and molecular light emerges as a promising alternative to conventional AD treatment interventions.
Collapse
Affiliation(s)
- Shi Kuang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown, Boston, MA-02129, USA
| | - Biyue Zhu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown, Boston, MA-02129, USA
| | - Jing Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown, Boston, MA-02129, USA
| | - Fan Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown, Boston, MA-02129, USA
| | - Bo Wu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown, Boston, MA-02129, USA
| | - Weihua Ding
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA-02129, USA
| | - Liuyue Yang
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA-02129, USA
| | - Shiqian Shen
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA-02129, USA
| | - Seven H Liang
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA-02114, USA
| | - Prasenjit Mondal
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA-02129, USA
| | - Mohanraja Kumar
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA-02139, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA-02129, USA
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA-02129, USA
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown, Boston, MA-02129, USA
| |
Collapse
|
28
|
Arfat Y, Zafar I, Sehgal SA, Ayaz M, Sajid M, Khan JM, Ahsan M, Rather MA, Khan AA, Alshehri JM, Akash S, Nepovimova E, Kuca K, Sharma R. In silico designing of multiepitope-based-peptide (MBP) vaccine against MAPK protein express for Alzheimer's disease in Zebrafish. Heliyon 2023; 9:e22204. [PMID: 38058625 PMCID: PMC10695983 DOI: 10.1016/j.heliyon.2023.e22204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/24/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023] Open
Abstract
Understanding the role of the mitogen-activated protein kinases (MAPKs) signalling pathway is essential in advancing treatments for neurodegenerative disorders like Alzheimer's. In this study, we investigate in-silico techniques involving computer-based methods to extract the MAPK1 sequence. Our applied methods enable us to analyze the protein's structure, evaluate its properties, establish its evolutionary relationships, and assess its prevalence in populations. We also predict epitopes, assess their ability to trigger immune responses, and check for allergenicity using advanced computational tools to understand their immunological properties comprehensively. We apply virtual screening, docking, and structure modelling to identify promising drug candidates, analyze their interactions, and enhance drug design processes. We identified a total of 30 cell-targeting molecules against the MAPK1 protein, where we selected top 10 CTL epitopes (PAGGGPNPG, GGGPNPGSG, SAPAGGGPN, AVSAPAGGG, AGGGPNPGS, ATAAVSAPA, TAAVSAPAG, ENIIGINDI, INDIIRTPT, and NDIIRTPTI) for further evaluation to determine their potential efficacy, safety, and suitability for vaccine design based on strong binding potential. The potential to cover a large portion of the world's population with these vaccines is substantial-88.5 % for one type and 99.99 % for another. In exploring the molecular docking analyses, we examined a library of compounds from the ZINC database. Among them, we identified twelve compounds with the lowest binding energy. Critical residues in the MAPK1 protein, such as VAL48, LYS63, CYS175, ASP176, LYS160, ALA61, LEU165, TYR45, SER162, ARG33, PRO365, PHE363, ILE40, ASN163, and GLU42, are pivotal for interactions with these compounds. Our result suggests that these compounds could influence the protein's behaviour. Moreover, our docking analyses revealed that the predicted peptides have a strong affinity for the MAPK1 protein. These peptides form stable complexes, indicating their potential as potent inhibitors. This study contributes to the identification of new drug compounds and the screening of their desired properties. These compounds could potentially help reduce the excessive activity of MAPK1, which is linked to Alzheimer's disease.
Collapse
Affiliation(s)
- Yasir Arfat
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, 56300, Pakistan
| | - Imran Zafar
- Department of Bioinformatics and Computational Biology, Virtual University, Punjab, 54700, Pakistan
| | - Sheikh Arslan Sehgal
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Mazhar Ayaz
- Department of Parasitology, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Muhammad Sajid
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, 56300, Pakistan
| | - Jamal Muhammad Khan
- Department of Parasitology, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Muhammad Ahsan
- Department of Environmental Sciences, Institute of Environmental and Agricultural Sciences, University of Okara, Okara, 56300, Pakistan
| | - Mohd Ashraf Rather
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries, Rangil- Gandarbal (SKAUST-K), India
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Jamilah M. Alshehri
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International, University, Dhaka, Bangladesh
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, 50 003, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, 50 003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Rohit Sharma
- Department of Rasashastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
29
|
Du DX, Khang NHD, Tri NH, Nam PC, Thong NM. Exploring the Multitarget Activity of Wedelolactone against Alzheimer's Disease: Insights from In Silico Study. ACS OMEGA 2023; 8:15031-15040. [PMID: 37151498 PMCID: PMC10157682 DOI: 10.1021/acsomega.2c08014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/07/2023] [Indexed: 05/09/2023]
Abstract
In this study, Wedelolactone's multitarget activity against Alzheimer's disease was examined using density functional theory and molecular docking techniques. At physiological pH, the pK a and molar fractions have been estimated. The most likely relative rate constants of two radical scavenger mechanisms are formal hydrogen transfer in a lipid environment and single-electron transfer in a water solvent. Compared to Trolox (k overall = 8.96 × 104 M-1 s-1), Wedelolactone (k overall = 4.26 × 109 M-1 s-1) is more efficient in scavenging the HOO• radical in an aqueous environment. The chelation capacity of metals was investigated by examining the complexation of the Cu(II) ion at various coordination positions and calculating the complexation kinetic constants. Furthermore, molecular docking simulations showed that the known forms of Wedelolactone at physiological pH effectively inhibited the AChE and BChE enzymes by comparing their activity to that of tacrine (control). Wedelolactone is a promising drug candidate for Alzheimer's disease therapy in light of these findings.
Collapse
Affiliation(s)
- Dang Xuan Du
- Sai
Gon University, 273 An Duong Vuong Street, Ho Chi Minh 700000, Vietnam
| | | | - Nguyen Huu Tri
- Sai
Gon University, 273 An Duong Vuong Street, Ho Chi Minh 700000, Vietnam
| | - Pham Cam Nam
- The
University of Danang - University of Science and Technology, Danang 550000, Vietnam
| | - Nguyen Minh Thong
- The
University of Danang - Campus in Kon Tum, 704 Phan Dinh Phung, Kon
Tum 580000, Vietnam
| |
Collapse
|
30
|
Khezri MR, Ghasemnejad-Berenji M. Gut microbiota and circadian rhythm in Alzheimer's disease pathophysiology: a review and hypothesis on their association. NPJ AGING 2023; 9:9. [PMID: 37130863 PMCID: PMC10154390 DOI: 10.1038/s41514-023-00104-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 03/15/2023] [Indexed: 05/04/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and the leading cause of dementia worldwide. Different pathologic changes have been introduced to be involved in its progression. Although amyloid-β (Aβ) deposition and tau hyperphosphorylation and aggregation are mainly considered the main characterizations of AD, several other processes are involved. In recent years, several other changes, including alterations in gut microbiota proportion and circadian rhythms, have been noticed due to their role in AD progression. However, the exact mechanism indicating the association between circadian rhythms and gut microbiota abundance has not been investigated yet. This paper aims to review the role of gut microbiota and circadian rhythm in AD pathophysiology and introduces a hypothesis to explain their association.
Collapse
Affiliation(s)
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
- Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
31
|
Singh PK, Donnenberg MS. High throughput and targeted screens for prepilin peptidase inhibitors do not identify common inhibitors of eukaryotic gamma-secretase. Expert Opin Drug Discov 2023; 18:563-573. [PMID: 37073444 PMCID: PMC11558661 DOI: 10.1080/17460441.2023.2203480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/12/2023] [Indexed: 04/20/2023]
Abstract
INTRODUCTION Prepilin peptidases (PPP) are essential enzymes for the biogenesis of important virulence factors, such as type IV pili (T4P), type II secretion systems, and other T4P-related systems of bacteria and archaea. PPP inhibitors could be valuable pharmaceuticals, but only a few have been reported. Interestingly, PPP share similarities with presenilin enzymes from the gamma-secretase protease complex, which are linked to Alzheimer's disease. Numerous gamma-secretase inhibitors have been reported, and some have entered clinical trials, but none has been tested against PPP. OBJECTIVE The objective of this study is to develop a high-throughput screening (HTS) method to search for inhibitors of PPP from various chemical libraries and reported gamma-secretase inhibitors. METHOD More than 15,000 diverse compounds, including 13 reported gamma-secretase inhibitors and other reported peptidase inhibitors, were screened to identify potential PPP inhibitors. RESULTS The authors developed a novel screening method and screened 15,869 compounds. However, the screening did not identify a PPP inhibitor. Nevertheless, the study suggests that gamma-secretase is sufficiently different from PPP that specific inhibitors may exist in a larger chemical space. CONCLUSION The authors believe that the HTS method that they describe has numerous advantages and encourage others to consider its application in the search for PPP inhibitors.
Collapse
Affiliation(s)
- Pradip Kumar Singh
- Department of Internal Medicine, Virginia Commonwealth University, Sanger Hall, Richmond, VA, USA
| | - Michael S Donnenberg
- Department of Internal Medicine, Virginia Commonwealth University, Sanger Hall, Richmond, VA, USA
| |
Collapse
|
32
|
Li K, Wang Z. lncRNA NEAT1: Key player in neurodegenerative diseases. Ageing Res Rev 2023; 86:101878. [PMID: 36738893 DOI: 10.1016/j.arr.2023.101878] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Neurodegenerative diseases are the most common causes of disability worldwide. Given their high prevalence, devastating symptoms, and lack of definitive diagnostic tests, there is an urgent need to identify potential biomarkers and new therapeutic targets. Long non-coding RNAs (lncRNAs) have recently emerged as powerful regulatory molecules in neurodegenerative diseases. Among them, lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) has been reported to be upregulated in Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). However, whether this is part of a protective or harmful mechanism is still unclear. This review summarizes our current knowledge of the role of NEAT1 in neurodegenerative diseases and its association with the characteristic aggregation of misfolded proteins: amyloid-β and tau in AD, α-synuclein in PD, mutant huntingtin in HD, and TAR DNA-binding protein-43 fused in sarcoma/translocated in liposarcoma in ALS. The aim of this review is to stimulate further research on more precise and effective treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Kun Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Ziqiang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China.
| |
Collapse
|
33
|
Hu Y, Zhou X, Wang L, Gu J, Zuo Y, Zhao L, Lu W, Yu Y. A liposome-based aptasensor integrated with competitive reaction enabling portable and electrochemical detection of Aβ oligomer. Biosens Bioelectron 2023; 225:115108. [PMID: 36709587 DOI: 10.1016/j.bios.2023.115108] [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: 10/01/2022] [Revised: 01/09/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
Aggregation of β-amyloid (Aβ) were considered as a typical pathological feature of Alzheimer's disease (AD). Extensive studies have verified that soluble Aβ oligomers (AβO) were more toxic to neurons than plaques. Herein, in this work, a glucose entrapped liposome-based portable aptasensor was fabricated for recognizing and interacting with AβO by specific aptamer on liposome (G-Lip-Apt). Then, a single strand DNA, designed to be partially complementary to AβO aptamer, was modified on amino-functionalized Fe3O4@SiO2 to obtain a magnetic nanocomposite (Fe3O4@SiO2/NH2-DNA). In the presence of AβO, the specific recognition between AβO and its aptamer on G-Lip-Apt made AβO bounded with G-Lip-Apt. With subsequent introduction of Fe3O4@SiO2/NH2-DNA, the unreacted G-Lip-Apt was further linked with Fe3O4@SiO2/NH2-DNA by double stranded complementary pairing interaction. Along with the addition of TritonX-100 into the formed G-Lip-Apt/Fe3O4@SiO2/NH2-DNA complex, the encapsulated glucose was released from liposome and then measured by a personal glucose meter (PGM). Good linear correlation was acquired over concentration of 5.0-1000 nM and the limit of detection (LOD) was calculated to be 2.27 nM for AβO. The developed portable electrochemical strategy integrated magnetic separation, competitive reaction and point of care test (POCT) to achieve high sensitivity, selectivity and accuracy, therefore enabled it successfully applied to the analysis of AβO in the hippocampus and cortex of APP/PS1 transgenic AD mice.
Collapse
Affiliation(s)
- Yuanyuan Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Xinguang Zhou
- Shenzhen NTEK Testing Technology Co., Ltd., Shenzhen, 518000, Guangdong, PR China
| | - Liming Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Jinyu Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Yingchun Zuo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Li Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Wenwen Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, PR China.
| |
Collapse
|
34
|
Cheng Y, He CY, Tian DY, Chen SH, Ren JR, Sun HL, Xu MY, Tan CR, Fan DY, Jian JM, Sun PY, Zeng GH, Shen YY, Shi AY, Jin WS, Bu XL, Liu HM, Xu YM, Wang J, Wang YJ. Physiological β-amyloid clearance by the liver and its therapeutic potential for Alzheimer's disease. Acta Neuropathol 2023; 145:717-731. [PMID: 36964213 DOI: 10.1007/s00401-023-02559-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/26/2023]
Abstract
Cerebral amyloid-β (Aβ) accumulation due to impaired Aβ clearance is a pivotal event in the pathogenesis of Alzheimer's disease (AD). Considerable brain-derived Aβ is cleared via transporting to the periphery. The liver is the largest organ responsible for the clearance of metabolites in the periphery. Whether the liver physiologically clears circulating Aβ and its therapeutic potential for AD remains unclear. Here, we found that about 13.9% of Aβ42 and 8.9% of Aβ40 were removed from the blood when flowing through the liver, and this capacity was decreased with Aβ receptor LRP-1 expression down-regulated in hepatocytes in the aged animals. Partial blockage of hepatic blood flow increased Aβ levels in both blood and brain interstitial fluid. The chronic decline in hepatic Aβ clearance via LRP-1 knockdown specific in hepatocytes aggravated cerebral Aβ burden and cognitive deficits, while enhancing hepatic Aβ clearance via LRP-1 overexpression attenuated cerebral Aβ deposition and cognitive impairments in APP/PS1 mice. Our findings demonstrate that the liver physiologically clears blood Aβ and regulates brain Aβ levels, suggesting that a decline of hepatic Aβ clearance during aging could be involved in AD development, and hepatic Aβ clearance is a novel therapeutic approach for AD.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Chen-Yang He
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, 610000, China
| | - Ding-Yuan Tian
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Si-Han Chen
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jun-Rong Ren
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Hao-Lun Sun
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Man-Yu Xu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Cheng-Rong Tan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Dong-Yu Fan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Jie-Ming Jian
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Pu-Yang Sun
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Ying-Ying Shen
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - An-Yu Shi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Wang-Sheng Jin
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Hong-Ming Liu
- Department of Hepatobiliary Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China.
| |
Collapse
|
35
|
Taha M, Rahim F, Zaman K, Anouar EH, Uddin N, Nawaz F, Sajid M, Khan KM, Shah AA, Wadood A, Rehman AU, Alhibshi AH. Synthesis, in vitro biological screening and docking study of benzo[ d]oxazole bis Schiff base derivatives as a potent anti-Alzheimer agent. J Biomol Struct Dyn 2023; 41:1649-1664. [PMID: 34989316 DOI: 10.1080/07391102.2021.2023640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We have synthesized benzo[d]oxazole derivatives (1-21) through a multistep reaction. Alteration in the structure of derivatives was brought in the last step via using various substituted aromatic aldehydes. In search of an anti-Alzheimer agent, all derivatives were evaluated against acetylcholinesterase and butyrylcholinesterase enzyme under positive control of standard drug donepezil (IC50 = 0.016 ± 0.12 and 4.5 ± 0.11 µM) respectively. In case of acetylcholinesterase enzyme inhibition, derivatives 8, 9 and 18 (IC50 = 0.50 ± 0.01, 0.90 ± 0.05 and 0.3 ± 0.05 µM) showed very promising inhibitory potentials. While in case of butyrylcholinesterase enzyme inhibition, most of the derivatives like 6, 8, 9, 13, 15, 18 and 19 (IC50 = 2.70 ± 0.10, 2.60 ± 0.10, 2.20 ± 0.10, 4.25 ± 0.10, 3.30 ± 0.10, 0.96 ± 0.05 and 3.20 ± 0.10 µM) displayed better inhibitory potential than donepezil. Moreover, derivative 18 is the most potent one among the series in both inhibitions. The binding interaction of derivatives with the active gorge of the enzyme was confirmed via a docking study. Furthermore, the binding interaction between derivatives and the active site of enzymes was correlated through the SAR study. Structures of all derivatives were confirmed through spectroscopic techniques such as 1H-NMR, 13C-NMR and HREI-MS, respectively.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Khalid Zaman
- Department of Chemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - El Hassane Anouar
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Nizam Uddin
- Department of Chemistry, University of Karachi, Karachi, Pakistan
| | - Faisal Nawaz
- Department of Chemistry, University of Wah, Wah Cantt, Pakistan
| | - Muhammad Sajid
- Department of Biochemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Adnan Ali Shah
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor 42300, Malaysia.,Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor 42300, Malaysia
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Ashfaq Ur Rehman
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Amani H Alhibshi
- Department of Neuroscience Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
36
|
Santoro A, Buonocore M, Grimaldi M, Napolitano E, D’Ursi AM. Monitoring the Conformational Changes of the Aβ(25-35) Peptide in SDS Micelles: A Matter of Time. Int J Mol Sci 2023; 24:ijms24020971. [PMID: 36674488 PMCID: PMC9867351 DOI: 10.3390/ijms24020971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/28/2022] [Accepted: 12/31/2022] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease is a neurodegenerative disease characterized by the formation of amyloid plaques constituted prevalently by amyloid peptides. Due to the well-known challenges related to the study in solution of these peptides, several membrane-mimicking systems such as micelle constituted by detergent-i.e., DPC and SDS-have been deeply investigated. Additionally, the strategy of studying short fragments instead of the full-length peptide turned out to be advantageous in exploring the structural properties of the different moieties in Aβ in order to reproduce its pathologic effects. Several studies reveal that among Aβ fragments, Aβ(25-35) is the shortest fragment able to reproduce the aggregation process. To enrich the structural data currently available, in the present work we decided to evaluate the conformational changes adopted by Aβ(25-35) in SDS combining CD and NMR spectroscopies at different times. From the solved structures, it emerges that Aβ(25-35) passes from an unordered conformation at the time of the constitution of the system to a more ordered and energetically favorable secondary structure at day 7, which is kept for 2 weeks. These preliminary data suggest that a relatively long time affects the kinetic in the aggregation process of Aβ(25-35) in a micellar system, favoring the stabilization and the formation of a soluble helix conformation.
Collapse
Affiliation(s)
- Angelo Santoro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
- Department of Pharmacy, Scuola di Specializzazione in Farmacia Ospedaliera, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Michela Buonocore
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
- Department of Veterinary Pathology, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy
| | - Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Enza Napolitano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Anna Maria D’Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
- Correspondence:
| |
Collapse
|
37
|
Abstract
Perturbation of cell membranes by amyloid β (Ab) peptide oligomers is one possible mechanism of cytotoxicity in Alzheimer's disease, but the structure of such Ab-membrane complexes is unknown. Here we examine the stability of several putative structures by implicit membrane and all-atom molecular dynamics simulations. The structures include (a) a variety of models proposed by other researchers in the past, (b) a heptameric β barrel determined by grafting the Ab sequence onto α-hemolysin, (c) a similar structure with modified strand orientation and turn location based on an experimental β-hairpin structure, (d) oligomers inserting C-terminal β hairpins into one leaflet of the bilayer, (e) oligomers forming parallel C-terminal β barrels, and (f) a helical hexamer made of C-terminal fragments. The α-hemolysin-grafted structure and its alternately oriented variant are stable in the membrane and form an aqueous pore. In contrast, the C-terminal parallel barrels are not stable, presumably due to excessive hydrophobicity of their inner surface. The helical hexamer also failed to stabilize an aqueous pore for the same reason. The C-terminal hairpin-inserting structures remain stably inserted but, again, do not form an aqueous pore. Our results suggest that only β-barrels inserting a combination of C-terminal and other residues can form stable aqueous pores.
Collapse
Affiliation(s)
- Aliasghar Sepehri
- Department of Chemistry, City College of New York, CUNY, 160 Convent Avenue, New York, New York10031, United States
| | - Themis Lazaridis
- Department of Chemistry, City College of New York, CUNY, 160 Convent Avenue, New York, New York10031, United States.,Graduate Programs in Chemistry, Biochemistry, and Physics, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, New York10016, United States
| |
Collapse
|
38
|
Li S, Huang Y, Yu L, Ji X, Wu J. Impact of the Cannabinoid System in Alzheimer's Disease. Curr Neuropharmacol 2023; 21:715-726. [PMID: 35105293 PMCID: PMC10207907 DOI: 10.2174/1570159x20666220201091006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 02/05/2023] Open
Abstract
Cannabinoids are compounds isolated from cannabis and are also widely present in both nervous and immune systems of animals. In recent years, with in-depth research on cannabinoids, their clinical medicinal value has been evaluated, and many exciting achievements have been continuously accumulating, especially in the field of neurodegenerative disease. Alzheimer's disease is the most common type of neurodegenerative disease that causes dementia and has become a global health problem that seriously impacts human health today. In this review, we discuss the therapeutic potential of cannabinoids for the treatment of Alzheimer's disease. How cannabinoids act on different endocannabinoid receptor subtypes to regulate Alzheimer's disease and the roles of the endocannabinoid system in Alzheimer's disease are outlined, and the underlying mechanisms are discussed. Finally, we summarize the most relevant opportunities of cannabinoid pharmacology related to Alzheimer's disease and discuss the potential usefulness of cannabinoids in the clinical treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Shuangtao Li
- Shantou University Medical College, Brain Function and Disease Laboratory, Shantou, #22 Road Xinling, Guangdong 515041, China
| | - Yuanbing Huang
- Department of Neurology, Yunfu People’s Hospital, Yunfu, Guangdong 527300, China
| | - Lijun Yu
- Shantou University Medical College, Brain Function and Disease Laboratory, Shantou, #22 Road Xinling, Guangdong 515041, China
| | - Xiaoyu Ji
- Department of Neurology, Yunfu People’s Hospital, Yunfu, Guangdong 527300, China
| | - Jie Wu
- Shantou University Medical College, Brain Function and Disease Laboratory, Shantou, #22 Road Xinling, Guangdong 515041, China
| |
Collapse
|
39
|
Bhatt S, Dhar AK, Samanta MK, Suttee A. Effects of Current Psychotropic Drugs on Inflammation and Immune System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:407-434. [PMID: 36949320 DOI: 10.1007/978-981-19-7376-5_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The immune system and inflammation are involved in the pathological progression of various psychiatric disorders such as depression or major depressive disorder (MDD), generalized anxiety disorder (GAD) or anxiety, schizophrenia, Alzheimer's disease (AD), and Huntington's disease. It is observed that levels of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and other markers are highly increased in the abovementioned disorders. The inflammation and immune component also lead to enhance the oxidative stress. The oxidative stress and increased production of reactive oxygen species (ROS) are considered as important factors that are involved in pathological progression of psychiatric disorders. Increase production of ROS is associated with excessive inflammation followed by cell necrosis and death. The psychotropic drugs are mainly work through modulations of neurotransmitter system. However, it is evident that inflammation and immune modulation are also having important role in the progression of psychiatric disorders. Rationale of the use of current psychotropic drugs is modulation of immune system by them. However, the effects of psychotropic drugs on the immune system and how these might contribute to their efficacy remain largely unclear. The drugs may act through modification of inflammation and related markers. The main purpose of this book chapter is to address the role of current psychotropic drugs on inflammation and immune system. Moreover, it will also address the role of inflammation in the progression of psychiatric disorders.
Collapse
Affiliation(s)
- Shvetank Bhatt
- School of Pharmacy, Dr. Vishwanath Karad MIT World Peace University, Pune, Maharashtra, India
- Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, India
| | | | | | - Ashish Suttee
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
40
|
Khatua P, Mondal S, Gupta M, Bandyopadhyay S. In Silico Studies to Predict the Role of Solvent in Guiding the Conformations of Intrinsically Disordered Peptides and Their Aggregated Protofilaments. ACS OMEGA 2022; 7:43337-43345. [PMID: 36506131 PMCID: PMC9730305 DOI: 10.1021/acsomega.2c06235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/08/2022] [Indexed: 06/17/2023]
Abstract
The formation of amyloids due to the self-assembly of intrinsically disordered proteins or peptides is a hallmark for different neurodegenerative diseases. For example, amyloids formed by the amyloid beta (Aβ) peptides are responsible for the most devastating neuropathological disease, namely, Alzheimer's disease, while aggregation of α-synuclein peptides causes the etiology of another neuropathological disease, Parkinson's disease. Characterization of the intermediates and the final amyloid formed during the aggregation process is, therefore, crucial for microscopic understanding of the origin behind such diseases, as well as for the development of proper therapeutics to combat those. However, most of the research activities reported in this area have been directed toward examining the early stages of the aggregation process, including probing the conformational characteristics of the responsible protein/peptide in the monomeric state or in small oligomeric forms. This is because the small soluble oligomers have been found to be more deleterious than the final insoluble amyloids. This review discusses some of the recent findings obtained from our simulation studies on Aβ and α-synuclein monomers and small preformed Aβ aggregates. A molecular-level insight of the aggregation process with a special emphasis on the role of water in inducing the aggregation process has been provided.
Collapse
Affiliation(s)
- Prabir Khatua
- Molecular
Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Souvik Mondal
- Molecular
Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Madhulika Gupta
- Department
of Chemistry and Chemical Biology, Indian
Institute of Technology (Indian School of Mines) Dhanbad, Jharkhand 826004, India
| | - Sanjoy Bandyopadhyay
- Molecular
Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
41
|
Sonar K, Mancera RL. Characterization of the Conformations of Amyloid Beta 42 in Solution That May Mediate Its Initial Hydrophobic Aggregation. J Phys Chem B 2022; 126:7916-7933. [PMID: 36179370 DOI: 10.1021/acs.jpcb.2c04743] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intrinsically disordered peptides, such as amyloid β42 (Aβ42), lack a well-defined structure in solution. Aβ42 can undergo abnormal aggregation and amyloidogenesis in the brain, forming fibrillar plaques, a hallmark of Alzheimer's disease. The insoluble fibrillar forms of Aβ42 exhibit well-defined, cross β-sheet structures at the molecular level and are less toxic than the soluble, intermediate disordered oligomeric forms. However, the mechanism of initial interaction of monomers and subsequent oligomerization is not well understood. The structural disorder of Aβ42 adds to the challenges of determining the structural properties of its monomers, making it difficult to understand the underlying molecular mechanism of pathogenic aggregation. Certain regions of Aβ42 are known to exhibit helical propensity in different physiological conditions. NMR spectroscopy has shown that the Aβ42 monomer at lower pH can adopt an α-helical conformation and as the pH is increased, the peptide switches to β-sheet conformation and aggregation occurs. CD spectroscopy studies of aggregation have shown the presence of an initial spike in the amount of α-helical content at the start of aggregation. Such an increase in α-helical content suggests a mechanism wherein the peptide can expose critical non-polar residues for interaction, leading to hydrophobic aggregation with other interacting peptides. We have used molecular dynamics simulations to characterize in detail the conformational landscape of monomeric Aβ42 in solution to identify molecular properties that may mediate the early stages of oligomerization. We hypothesized that conformations with α-helical structure have a higher probability of initiating aggregation because they increase the hydrophobicity of the peptide. Although random coil conformations were found to be the most dominant, as expected, α-helical conformations are thermodynamically accessible, more so than β-sheet conformations. Importantly, for the first time α-helical conformations are observed to increase the exposure of aromatic and hydrophobic residues to the aqueous solvent, favoring their hydrophobically driven interaction with other monomers to initiate aggregation. These findings constitute a first step toward characterizing the mechanism of formation of disordered, low-order oligomers of Aβ42.
Collapse
Affiliation(s)
- Krushna Sonar
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin Institute for Computation, Curtin University, P. O. Box U1987, Perth, Western Australia6845, Australia
| | - Ricardo L Mancera
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin Institute for Computation, Curtin University, P. O. Box U1987, Perth, Western Australia6845, Australia
| |
Collapse
|
42
|
Yue C, Feng S, Chen Y, Jing N. The therapeutic prospects and challenges of human neural stem cells for the treatment of Alzheimer's Disease. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:28. [PMID: 36050613 PMCID: PMC9437172 DOI: 10.1186/s13619-022-00128-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder associated with aging. Due to its insidious onset, protracted progression, and unclear pathogenesis, it is considered one of the most obscure and intractable brain disorders, and currently, there are no effective therapies for it. Convincing evidence indicates that the irreversible decline of cognitive abilities in patients coincides with the deterioration and degeneration of neurons and synapses in the AD brain. Human neural stem cells (NSCs) hold the potential to functionally replace lost neurons, reinforce impaired synaptic networks, and repair the damaged AD brain. They have therefore received extensive attention as a possible source of donor cells for cellular replacement therapies for AD. Here, we review the progress in NSC-based transplantation studies in animal models of AD and assess the therapeutic advantages and challenges of human NSCs as donor cells. We then formulate a promising transplantation approach for the treatment of human AD, which would help to explore the disease-modifying cellular therapeutic strategy for the treatment of human AD.
Collapse
Affiliation(s)
- Chunmei Yue
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
| | - Su Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou, 510005, China
| | - Yingying Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou, 510005, China.
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
43
|
Mays CE, Trinh THT, Telling G, Kang HE, Ryou C. Endoproteolysis of cellular prion protein by plasmin hinders propagation of prions. Front Mol Neurosci 2022; 15:990136. [PMID: 36117913 PMCID: PMC9478470 DOI: 10.3389/fnmol.2022.990136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023] Open
Abstract
Many questions surround the underlying mechanism for the differential metabolic processing observed for the prion protein (PrP) in healthy and prion-infected mammals. Foremost, the physiological α-cleavage of PrP interrupts a region critical for both toxicity and conversion of cellular PrP (PrP C ) into its misfolded pathogenic isoform (PrP Sc ) by generating a glycosylphosphatidylinositol (GPI)-anchored C1 fragment. During prion diseases, alternative β-cleavage of PrP becomes prominent, producing a GPI-anchored C2 fragment with this particular region intact. It remains unexplored whether physical up-regulation of α-cleavage can inhibit disease progression. Furthermore, several pieces of evidence indicate that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 play a much smaller role in the α-cleavage of PrP C than originally believed, thus presenting the need to identify the primary protease(s) responsible. For this purpose, we characterized the ability of plasmin to perform PrP α-cleavage. Then, we conducted functional assays using protein misfolding cyclic amplification (PMCA) and prion-infected cell lines to clarify the role of plasmin-mediated α-cleavage during prion propagation. Here, we demonstrated an inhibitory role of plasmin for PrP Sc formation through PrP α-cleavage that increased C1 fragments resulting in reduced prion conversion compared with non-treated PMCA and cell cultures. The reduction of prion infectious titer in the bioassay of plasmin-treated PMCA material also supported the inhibitory role of plasmin on PrP Sc replication. Our results suggest that plasmin-mediated endoproteolytic cleavage of PrP may be an important event to prevent prion propagation.
Collapse
Affiliation(s)
- Charles E. Mays
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Trang H. T. Trinh
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, South Korea,Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Glenn Telling
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States,Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Hae-Eun Kang
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States,Reference Laboratory for Chronic Wasting Disease (CWD), Foreign Animal Disease Division, Animal and Plant Quarantine Agency, Gimcheon, South Korea,Hae-Eun Kang,
| | - Chongsuk Ryou
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, United States,Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, South Korea,Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea,Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States,*Correspondence: Chongsuk Ryou,
| |
Collapse
|
44
|
Cho Y, Bae HG, Okun E, Arumugam TV, Jo DG. Physiology and pharmacology of amyloid precursor protein. Pharmacol Ther 2022; 235:108122. [PMID: 35114285 DOI: 10.1016/j.pharmthera.2022.108122] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
Abstract
Amyloid precursor protein (APP) is an evolutionarily conserved transmembrane protein and a well-characterized precursor protein of amyloid-beta (Aβ) peptides, which accumulate in the brains of individuals with Alzheimer's disease (AD)-related pathologies. Aβ has been extensively investigated since the amyloid hypothesis in AD was proposed. Besides Aβ, previous studies on APP and its proteolytic cleavage products have suggested their diverse pathological and physiological functions. However, their roles still have not been thoroughly understood. In this review, we extensively discuss the evolutionarily-conserved biology of APP, including its structure and processing pathway, as well as recent findings on the physiological roles of APP and its fragments in the central nervous system and peripheral nervous system. We have also elaborated upon the current status of APP-targeted therapeutic approaches for AD treatment by discussing inhibitors of several proteases participating in APP processing, including α-, β-, and γ-secretases. Finally, we have highlighted the future perspectives pertaining to further research and the potential clinical role of APP.
Collapse
Affiliation(s)
- Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Han-Gyu Bae
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Pauld Feder Laboratory on Alzheimer's Disease Research, Israel
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea; School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia.
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea; Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, South Korea; Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
45
|
Kuragano M, Yamanaka S, Tokuraku K. Kinetics of amyloid accumulation in physiological viscosity. Colloids Surf B Biointerfaces 2022; 214:112449. [PMID: 35306343 DOI: 10.1016/j.colsurfb.2022.112449] [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: 10/26/2021] [Revised: 02/18/2022] [Accepted: 03/07/2022] [Indexed: 11/24/2022]
Abstract
Abnormal aggregation and accumulation of misfolded proteins are involved in the development of various forms of amyloidosis. Aggregates that accumulate in organs induce an inflammatory response and cytotoxicity, and lead to organ failure. Although protein accumulation around an affected area in the body is an important stage that is directly linked to the mechanism of pathogenesis, the kinetics of the accumulation of protein that precipitates while assembling is not well understood because 3D tracking of proteins in solution is difficult. Here, we analyzed the process of aggregation and accumulation of amyloid β (Aβ), which causes the development of Alzheimer's disease (AD), by real-time 3D imaging under physiological conditions using a quantum dot nanoprobe that we previously developed. 3D observations demonstrated that Aβ aggregates with a diameter of several μm emerged in phosphate-buffered saline, gathered in a spiral-like step, and exhibited a mesh-like structure. Additionally, we found that the amount and size of aggregates decreased dramatically in 40% glycerol solution, mimicking the viscosity of human blood. We confirmed that fibrils in 40% glycerol exhibited an extremely short and tangled morphology and formed dense aggregates. Furthermore, numerical calculations revealed that several decades are required to fully develop the settling velocity and diameter of Aβ aggregates in physiological conditions. This time span is consistent with the actual symptom progression of AD.
Collapse
Affiliation(s)
- Masahiro Kuragano
- Graduate School of Engineering, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Shinya Yamanaka
- Graduate School of Engineering, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Kiyotaka Tokuraku
- Graduate School of Engineering, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan.
| |
Collapse
|
46
|
Cheng K, Huang C, Hsieh T, Chiang H. Disrupted cellular calcium homeostasis is responsible for Aβ‐induced learning and memory damage and lifespan shortening in a model of Aβ transgenic fly. IUBMB Life 2022; 74:754-762. [DOI: 10.1002/iub.2621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 04/11/2022] [Indexed: 12/27/2022]
Affiliation(s)
- Kuan‐Chung Cheng
- Department of Pharmacology, College of Medicine National Cheng‐Kung University Tainan Taiwan
- Institute of Basic Medical Sciences, College of Medicine National Cheng‐Kung University Tainan Taiwan
| | - Chih‐Yuan Huang
- Division of Nephrology, Department of Internal Medicine Ditmanson Medical Foundation Chia‐Yi Christian Hospital Chiayi Taiwan
- Department of Sport Management, College of Recreation and Health Management Chia Nan University of Pharmacy and Science Tainan Taiwan
| | - Tsung‐Chi Hsieh
- Department of Pharmacology, College of Medicine National Cheng‐Kung University Tainan Taiwan
- Institute of Basic Medical Sciences, College of Medicine National Cheng‐Kung University Tainan Taiwan
- Brain Research Center National Tsing Hua University Hsinchu City Taiwan
| | - Hsueh‐Cheng Chiang
- Department of Pharmacology, College of Medicine National Cheng‐Kung University Tainan Taiwan
- Institute of Basic Medical Sciences, College of Medicine National Cheng‐Kung University Tainan Taiwan
| |
Collapse
|
47
|
Qian X, Chen K, Chen L, Song H, Zhang Z. Presenilin is involved in larval-pupal metamorphosis development of Bombyx mori. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 109:e21855. [PMID: 34811799 DOI: 10.1002/arch.21855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 06/13/2023]
Abstract
Disruption of the presenilin (ps) genes are the major genetic cause of familial Alzheimer's disease. The silkworm, Bombyx mori (B. mori), is an important model insect. The ps homologue gene in B. mori was identified and characterized. However, the role of ps in B. mori was poorly understood. Here, we found that Bmps was ubiquitously expressed in all the tested tissues during metamorphosis. In the current study, loss-of-function analysis of Bmps was performed by the binary transgenic CRISPR/cas9 system. Compared with the wild type, the developmental time of ∆Bmps animals were significantly delayed. In addition, ∆Bmps showed abnormal appendage including antenna, leg, wing and eye during pupal and adult stages. RNA-seq analysis indicated that apoptosis and proliferation related pathways were affected in ∆Bmps. Moreover, the Hippo pathway was affected by Bmps depletion in brain and wing disc. Our results suggest that PS is essential for maintaining the dynamic balance of apoptosis and proliferation during metamorphosis.
Collapse
Affiliation(s)
- Xiaoran Qian
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Kai Chen
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu, China
| | - Lijuan Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Hongsheng Song
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Zhongjie Zhang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
48
|
Wang BY, Gu BC, Wang GJ, Yang YH, Wu CC. Detection of Amyloid-β(1–42) Aggregation With a Nanostructured Electrochemical Sandwich Immunoassay Biosensor. Front Bioeng Biotechnol 2022; 10:853947. [PMID: 35372290 PMCID: PMC8965719 DOI: 10.3389/fbioe.2022.853947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/25/2022] [Indexed: 11/30/2022] Open
Abstract
Amyloid-β(1–42) [Aβ(1–42)] oligomer accumulations are associated with physiologic alterations in the brains of individuals with Alzheimer’s disease. In this study, we demonstrate that a nanostructured gold electrode with deposited gold nanoparticles, induced via electrochemical impedance spectroscopy (EIS), may be used as an Aβ(1–42) conformation biosensor for the detection of Alzheimer’s disease. Monoclonal antibodies (12F4) were immobilized on self-assembled monolayers of the electrochemical sandwich immunoassay biosensor to capture Aβ(1–42) monomers and oligomers. Western blot and fluorescence microscopy analyses were performed to confirm the presence of Aβ(1–42) monomers and oligomers. EIS analysis with an equivalent circuit model was used to determine the concentrations of different Aβ(1–42) conformations in this study. We identified conformations of Aβ(1–42) monomers and Aβ(1–42) oligomers using probe antibodies (12F4) by employing EIS. RAβ(1−42) indicates the sum resistance of impedance measured during Aβ(1–42) immobilization. ΔR12F4 refers to the concentration of probe antibody (12F4) binding with Aβ(1–42). The concentration of Aβ(1–42) oligomer was defined as the percentage of Aβ(1–42) aggregation R12F4/RAβ(1−42). The experimental results show that the biosensor has high selectivity to differentiate Aβ(1–40) and Aβ(1–42) monomers and Aβ(1–42) oligomers and that it can detect Aβ(1–42) oligomer accurately. The linear detection range for Aβ(1–42) oligomers was between 10 pg/ml and 100 ng/ml. The limit of detection was estimated to be 113 fg/ml.
Collapse
Affiliation(s)
- Bing-Yu Wang
- Department of Mechanical Engineering, National Chung Hsing University, Taichung, Taiwan
| | - Bien-Chen Gu
- Department of Mechanical Engineering, National Chung Hsing University, Taichung, Taiwan
| | - Gou-Jen Wang
- Department of Mechanical Engineering, National Chung Hsing University, Taichung, Taiwan
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, Taiwan
| | - Yuan-Han Yang
- Department of and Master's Program in Neurology, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Che Wu
- Department of Mechanical Engineering, National Chung Hsing University, Taichung, Taiwan
- Innovation and Development Center of Sustainable Agriculture (IDCSA), National Chung Hsing University, Taichung, Taiwan
- Smart Sustainable New Agriculture Research Center (SMARTer), Taichung, Taiwan
| |
Collapse
|
49
|
Herbal Mixture of Carthamus tinctorius L. Seed and Taraxacum coreanum Attenuates Amyloid Beta-Induced Cognitive Dysfunction In Vivo. Foods 2022; 11:foods11020142. [PMID: 35053874 PMCID: PMC8774339 DOI: 10.3390/foods11020142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/24/2021] [Accepted: 01/04/2022] [Indexed: 12/31/2022] Open
Abstract
Deposition of amyloid-beta (Aβ) in the aging brain has been often observed and is thought to be a pathological feature of Alzheimer’s disease. The use of natural products for disease prevention and treatment is gaining attention worldwide. Carthamus tinctorius L. seed and Taraxacum coreanum have been used as traditional medicines in Asian countries, where they have been reported to exert anti-inflammatory and anti-oxidative effects. It has been demonstrated that the combination of C. tinctorius L. seed and T. coreanum has an effect on cognitive enhancement, indicating a ratio of 5:5 synergistically enhancing learning and memory abilities in comparison with a single treatment. Here, we aimed to investigate the protective effect of C. tinctorius L. seed and T. coreanum mixture (CT) at different concentrations on cognition in Aβ25-35-infused mice. CT-administered mice showed significant cognitive improvement in the T-maze, novel object recognition, and Morris water maze tests. Moreover, amyloidogenesis-related proteins, such as β-secretase and γ-secretase, were detected and their protein levels decreased after treatment with CT. Our study shows that CT attenuates cognitive dysfunction by improving learning and memory capability and regulating Aβ-related proteins in Aβ25-35-injected mice. These findings suggest that CT might be a candidate for functional food on cognitive improvement.
Collapse
|
50
|
In vitro neuroprotective effects of allicin on Alzheimer’s disease model of neuroblastoma cell line. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.1068336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|